Next Article in Journal
Uptake of Nanotitania by Gingival Epithelial Cells Promotes Inflammatory Response and Is Accelerated by Porphyromonas gingivalis Lipopolysaccharide: An In Vitro Study
Next Article in Special Issue
Antimicrobial Resistance and Whole-Genome Characterisation of High-Level Ciprofloxacin-Resistant Salmonella Enterica Serovar Kentucky ST 198 Strains Isolated from Human in Poland
Previous Article in Journal
Effects of Dihydrotanshinone I on Proliferation and Invasiveness of Paclitaxel-Resistant Anaplastic Thyroid Cancer Cells
Previous Article in Special Issue
Recent Advances in the Detection of Antibiotic and Multi-Drug Resistant Salmonella: An Update
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Coming from the Wild: Multidrug Resistant Opportunistic Pathogens Presenting a Primary, Not Human-Linked, Environmental Habitat

by
Fernando Sanz-García
,
Teresa Gil-Gil
,
Pablo Laborda
,
Luz E. Ochoa-Sánchez
,
José L. Martínez
* and
Sara Hernando-Amado
Centro Nacional de Biotecnología, CSIC, 28049 Madrid, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(15), 8080; https://doi.org/10.3390/ijms22158080
Submission received: 14 June 2021 / Revised: 22 July 2021 / Accepted: 24 July 2021 / Published: 28 July 2021
(This article belongs to the Special Issue Multi-Drug Resistant Bacteria—A Global Problem)

Abstract

:
The use and misuse of antibiotics have made antibiotic-resistant bacteria widespread nowadays, constituting one of the most relevant challenges for human health at present. Among these bacteria, opportunistic pathogens with an environmental, non-clinical, primary habitat stand as an increasing matter of concern at hospitals. These organisms usually present low susceptibility to antibiotics currently used for therapy. They are also proficient in acquiring increased resistance levels, a situation that limits the therapeutic options for treating the infections they cause. In this article, we analyse the most predominant opportunistic pathogens with an environmental origin, focusing on the mechanisms of antibiotic resistance they present. Further, we discuss the functions, beyond antibiotic resistance, that these determinants may have in the natural ecosystems that these bacteria usually colonize. Given the capacity of these organisms for colonizing different habitats, from clinical settings to natural environments, and for infecting different hosts, from plants to humans, deciphering their population structure, their mechanisms of resistance and the role that these mechanisms may play in natural ecosystems is of relevance for understanding the dissemination of antibiotic resistance under a One-Health point of view.

1. Introduction

Bacterial organisms causing human infections can be divided into two categories; those that infect healthy people, and those that mainly infect people with underlying diseases, immunosuppressed or debilitated. While the former are relevant both in the community and in the hospitals, the latter have been dubbed opportunistic pathogens and are primarily a hospital problem [1]. Opportunistic pathogens have historically originated from human commensal bacteria. Indeed, in the seminal paper that led to the search of antibiotic producers in soils, the main reason for such screening was that, despite the soil being a sink for organisms infecting humans, “one hardly thinks of the soil as a source of epidemics” [2]. Nevertheless, in the last decades, an increased prevalence of opportunistic pathogens with an environmental origin, most of them non-fermentative Gram-negative bacteria [3], has been reported [4]. Most of these pathogens present low susceptibility to antibiotics currently used in therapy, suggesting that the enrichment of these pathogens at hospitals can result from the selection pressure exerted by antibiotics used for treating infectious diseases [5]. Actually, one of the risk factors for being infected by these pathogens is previous antibiotic treatment with broad-spectrum antibiotics. As opposed to what Waksman and Woodruff stated in 1940 [2], it is now evident that natural environments encompass an undefined reservoir of bacterial species, some of which have the potential to infect humans. These infections mainly occur in immunodeficient people and patients with underlying diseases. This fact suggests that, beyond the existence of specific lineages that have evolved towards virulence, the main reason behind infection by this type of opportunistic pathogens is the health status of the infected patient. Indeed, for most of the opportunistic pathogens herein reviewed, there are not clear genomic differences between environmental and clinical isolates. This factor does not mean that epidemic clones are absent, but rather that those clones, more frequently involved in outbreaks at hospitals, are also present in natural ecosystems. Further, the fact that most of their virulence determinants and several antibiotic resistance genes (ARGs) are usually present in their core genomes supports that these elements have evolved to deal with functions other than infecting humans in the natural habitats that these microorganisms colonize. In the present article, we review the most relevant current information on these pathogens, with a particular emphasis on their mechanisms of antibiotic resistance (AR). It is important to notice that, besides being the primary habitat (i.e., the origin) of some opportunistic pathogens, natural ecosystems are the places where all human bacteria, pathogens and commensals end up, along with the ARGs they carry [6,7,8]. While environmental antibiotic-resistant organisms, such as Pseudomonas aeruginosa or Burkholderia cepacia, regularly colonize environmental habitats, other pathogens with relevance for the dissemination of resistance, such as Escherichia coli, Enterococcus or Klebsiella pneumoniae, are part of human-linked microbiomes; their finding in a natural ecosystem is considered a sign of anthropogenic pollution [9,10], to the extent that it has been stated that resistant organisms detected in wastewater treatment plants should reflect the overall resistome of the human populations they serve [11,12,13,14]. Certainly, upon such pollution, natural ecosystems can be drivers for the evolution and spread of AR in any human pathogen [6,15]; however, in the current review, we focus just on those organisms that present a bona fide, non-clinical, environmental primary habitat, where they have evolved [16,17] before causing human infections.

2. Pseudomonas aeruginosa

P. aeruginosa is a Gram-negative, rod-shaped, non-fermentative, facultative anaerobic bacterium able to colonize a wide range of different habitats due to its high metabolic versatility and broad capacity of adaptation to fluctuating environments [18]. Its presence has been described in soil [19], crude oil [20] or different aquatic systems such as wastewater [21], freshwater and seawater environments [22], being found among the most frequent locations those closely related to human activities [23].
Considering a host as an environment colonizable by bacteria [16], the ability of P. aeruginosa to cause infections in a large range of hosts may also be used to exemplify its high adaptability and ubiquitous distribution. P. aeruginosa infections have been reported in plants [24], animals -such as insects [25], nematodes [26], fishes [27] or mammals [28], including humans [29]- or even amoebas, such as Dictyostelium discoideum [30]. However, despite the wide distribution of P. aeruginosa, several studies have indicated that there are no specific clones associated with specific habitats and that environmental and clinical isolates are indistinguishable. In addition, there is a consensus about the non-clonal epidemic nature of P. aeruginosa population structure [31].
P. aeruginosa is one of the main causes of nosocomial infections, including acute respiratory diseases and bacteraemia [32]. Moreover, it can chronically infect immunocompromised people or patients with underlying diseases, such as chronic obstructive pulmonary disease (COPD) [33], cystic fibrosis (CF) [34], AIDS [35], cancer [36] or those presenting burn or surgical wounds [37], being that these infections are an utmost source of morbidity and mortality in intensive care units (ICUs).
The impact of P. aeruginosa on human health cannot be understood without taking into account the vast amount of virulence factors it possesses. Proteases, flagella, secretion systems, biofilm formation (which is particularly worrying when located in catheters, prosthesis or lungs [38,39,40]) or quorum sensing (QS) (the cell-cell signalling system that coordinates the expression of most of the said factors [41]), are elements that pave the way for infection and hamper therapies. Further, this microorganism exhibits low susceptibility to a great number of drugs [42], an issue that is dissected below. Overall, these features result in P. aeruginosa being subsumed into two bacterial ensembles, namely ESKAPE (acronym of Enterococcus faecium, Staphylococcus aureus, K. pneumoniae, Acinetobacter baumannii, P. aeruginosa, and Enterobacter spp.) and TOTEM (TOp TEn resistant Microorganisms), which include the currently most relevant multidrug-resistant human pathogens [43,44].
P. aeruginosa infections are frequently treated with aminoglycosides, especially tobramycin, as well as with cephalosporines or β-lactam/β-lactamase-inhibitor combinations, such as piperacillin/tazobactam or ceftazidime/avibactam [32]. Besides, fluoroquinolones (ciprofloxacin), polymyxins, fosfomycin, aztreonam and carbapenems are also antibiotics of choice, which usage depends on the characteristics of the infection [45]. Nevertheless, the aforementioned intrinsic resistance of this pathogen to some antibiotics has compelled many to search for novel β-lactam/β-lactamase inhibitors, like imipenem/relebactam [46], or the development of new antimicrobial compounds, such as plazomicin, murepavadin or doripenem [47]. It is also important to remark that non-antibiotic therapies have been delved into, in order to counteract treatment failure when resistance to classical drugs emerges. Among these strategies, anti-virulence compounds, efflux pump inhibitors and permeabilizing membrane compounds (co-administered with antibiotics or on their own) stand out as the most promising alternatives [48,49,50]. In addition, evolution-based approaches that exploit phenotypic convergence and negative hysteresis phenomena are currently being investigated to fight P. aeruginosa and other human pathogen infections [51,52,53,54].
The above-stated high intrinsic resistance to antibiotics of P. aeruginosa (Figure 1) is due to its low outer membrane permeability [55], the production of antibiotic-modifying enzymes [56], and the large stock of multidrug resistance (MDR) efflux pumps it harbours [47]. Concerning the latter, there are 12 Resistance Nodulation Division (RND) family members that have been ascribed to this bacterium; among them, MexAB-OprM, MexXY-OprM, MexCD-OprJ and MexEF-OprN are of significant interest, given their known role in clinical settings [57]. The first two are the ones that have been shown to contribute to intrinsic resistance, but every system is able to extrude a wide range of antimicrobial agents (Table 1). Regarding antibiotic-modifying enzymes, P. aeruginosa can resort to its inherent assortment of β-lactamases, making AmpC the most noteworthy [58], and aminoglycoside-modifying enzymes, namely aminoglycoside acetyltransferases, phosphotransferases and nucleotidyltransferases [59]. Additionally, it is worth emphasizing that its intrinsic resistome does not only consist of classical resistance determinants, but it may encompass basic components of bacterial physiology [60]. For example, Crc is a global regulator of carbon metabolism whose inactivation entails an increased susceptibility to several antimicrobials in P. aeruginosa [61]. This situation agrees with the notion that the ancestral, physiological function of intrinsic resistance determinants of pathogens with an environmental origin goes beyond counteracting the activity of antimicrobial agents currently used in therapy. As previously discussed [42], interfering with the effectiveness of antibiotics is a novel functional role of these determinants, promoted by the current antibiotic era.
In addition to intrinsic AR, increased resistance levels may also be acquired by chromosomal mutations that boost the expression of the above-described determinants (Figure 1), a situation that frequently takes place during chronic infections [62]. For instance, mutations in genes encoding regulators of MDR efflux pumps can lead to the overexpression of the latter and, as a consequence, a more efficient extrusion of drugs. In the case of mexAB-oprM, overexpression may be due to mutations in the gene that encodes its local repressor, MexR, or in genes encoding other secondary regulators, NalC or NalD, events that have been reported in vivo [63,64]. Similarly, mexCD-oprJ and mexXY-oprM overexpression could be driven by spontaneous mutations in genes encoding their repressors, NfxB and MexZ, respectively [63,65]. Concerning β-lactamases, indirect or direct repressors of ampC expression, AmpR or AmpD, are commonly found mutated in β-lactam resistant strains of P. aeruginosa, presenting an enhanced β-lactamase activity and, consequently, resistance to β-lactams [58].
Besides, mutations can be selected in genes that encode the resistance determinants themselves. In the case of MDR efflux pumps, it has been published that amino acid changes in MexY, a subunit of the MexXY-OprM pump, may optimize antibiotic recognition site and, hence, improve drug efflux [66]. Regarding enzymes, cephalosporinase AmpC variants are able to extend their substrate spectrum, becoming capable of hydrolyzing carbapenems [67] or recent β-lactam/β-lactamase inhibitor combinations [68].
Beyond modifications in resistance determinants, the mutational resistome of P. aeruginosa is still more multifarious. Some examples of the versatile mutational resistome of this opportunistic pathogen are the mutations in genes involved in the peptidoglycan recycling pathway, as mpl or dacB, which raise β-lactamase activity [69]; in genes that encode drug targets, as gyrA or gyrB, which foster quinolone resistance [70]; in genes encoding Penicillin Binding Proteins (PBPs), i.e., PBP3, which cause β-lactams resistance [71]; or even in loci that do not seem to be associated with AR, as pilQ, a gene that codes for a Type IV pili protein, which can give rise to resistance against various antipseudomonal agents [72].
Alternatively, this microorganism can acquire ARGs through Horizontal Gene Transfer (HGT) [47]. These genes can locate in integrative and conjugative elements (ICEs), plasmids, integrons, transposons or prophages (Figure 1), and they can be transferred by different mechanisms [73]. As expected, there is plethora of examples of ARG acquisition by P. aeruginosa through HGT, with ICEs and plasmids being the most usual ARGs carriers [74]. In this sense, an extensive miscellany of β-lactamases, aminoglycoside and fluoroquinolone-modifying enzymes have routinely been detected in these vectors [75,76,77,78], either alone or accompanied by large arrays of ARGs [79]. Correspondingly, integrons, which do not transfer independently, but are gene-recruiting elements, may also harbour metallo β-lactamases (MBLs) (i.e., carbapenemases) [80], aminoglycoside-modifying enzymes [81], or both [82], among other resistance mechanisms [83]. Phage particles containing ARGs have been found in the lungs of CF patients suffering chronic infections by P. aeruginosa [84]; in addition, a composite phage-like plasmid carrying the β-lactamase-encoding gene blaKPC-2 has been found in a carbapenem-resistant P. aeruginosa isolate [85]. However, a deep understanding of the relevance of prophages in disseminating ARGs in P. aeruginosa requires further studies. In conclusion, this pathogen wields an astonishing range of alternatives to achieve AR.
Lastly, from a One-Health perspective, it must be noted that P. aeruginosa ubiquity in nature aggravates the problem of AR. This bacterium has a broad sub-lethal selective window to different antibiotics, under which resistant mutants may arise [86,87]. This matter becomes more alarming since substantial concentrations of drugs (i.e., 31 or 61 mg/L of ciprofloxacin or tetracycline, respectively) have been detected in habitats that P. aeruginosa can colonize [23,88], besides clinical settings. Further, aquatic ecosystems have been suggested as reservoirs and sources of ARGs, usually carried on plasmids, a situation that may play a critical role in the propagation of antimicrobial resistance among P. aeruginosa strains [89].
Besides intrinsic and genetically acquired, stable AR, the resistance phenotype can be acquired transiently, without the need for genetic changes. Transient resistance can be achieved by the bulk of the population, as happens in the case of biofilms [90]. The capacity of P. aeruginosa to form a biofilm within a host–which contributes to its ability to inhabit diverse ecological niches- impedes phagocytosis and diminishes the efficiency of antimicrobial treatments, sometimes provoking chronic and persistent infections in host tissues or prosthetic devices [39]. Transient resistance can also be developed just by a bacterial subpopulation, a situation dubbed persistence, which is defined as the ability of a part of the bacterial population to survive under an antimicrobial treatment without acquiring genetic changes conferring resistance [91]. In P. aeruginosa, persistence is encountered under nutrient limiting conditions [92], as is the presence of QS signaling molecules [93].
One explanation for the increased antimicrobial resistance of biofilm-growing P. aeruginosa is the presence of cells with a slow-growing metabolic state in some parts of the biofilm, which constitute a subpopulation of persisters [94]. Other reasons for an increased transient resistance are a more difficult diffusion of compounds due to the complex structure of the biofilm [90] or the presence of elements that reduce the activity of antimicrobials within the biofilm, such as glycerophosphorylated β-(1, 3)-glucans or cyclic-β-(1, 3)-glucans, which sequester aminoglycoside antibiotics [95]. In addition, changes in the expression levels of ARGs as mexAB-oprM and mexCD-oprJ during the biofilms state of growth are also of relevance [96,97].
Besides persistence and the formation of recalcitrant biofilm structures, changes in the expression of AR determinants due to specific signals and/or conditions are of relevance for developing transient resistance. Indeed, this scenario may be encountered during infection, thus compromising the efficacy of antipseudomonal treatments [98]. For instance, the presence of the inducible β-lactamase AmpC in P. aeruginosa, the expression of which is enhanced by some β-lactam antibiotics, may lead to treatment failure due to transient β-lactam resistance [99]. Changes in the permeability of P. aeruginosa associated with magnesium limiting conditions transitorily reduce the negative charge of the cell surface through an up-regulation of an LPS modification operon, driving to enhanced resistance in positively charged antimicrobials, like cationic antimicrobial peptides or polymyxin B [100,101].
The role of MDR efflux pumps regarding P. aeruginosa transient AR should also be highlighted. Since these systems are involved in different key processes for bacterial physiology, tight regulatory control over their expression, dependent on environmental conditions, may be expected. Therefore, in some situations or in the presence of specific effectors, a temporary rise of the expression of efflux pump encoding genes is achieved [102]. Some of these inducing conditions may be found in clinical settings, allowing bacteria to resist an antimicrobial treatment through a transitory improved antibiotic extrusion capacity. For instance, expression of MexCD-OprJ efflux pump’s encoding genes is induced by molecules that P. aeruginosa may run into during an infection. Some of these molecules are disinfectants or anaesthetic agents (e.g., procaine or atropine), which induce quinolone resistance [103], as well as the human host defence peptide LL-37, which increases resistance towards quinolones and aminoglycosides [104].
Further, mexAB-oprM expression is induced under oxidative stress conditions [105] and by triclosan or pentachlorophenol [106]. Furthermore, nitrosative stress, chloramphenicol presence and contact with human airway epithelial cells are circumstances that trigger mexEF-oprN overexpression [107,108], supporting the idea that the MexEF-OprN efflux pump might contribute to P. aeruginosa transient resistance during lung infection. Another P. aeruginosa’s efflux pump, MexXY, may also contribute to transient AR in clinical settings since the expression of its encoding genes is induced under oxidative stress conditions or in the presence of antibiotics able to inhibit protein synthesis as aminoglycosides or tetracyclines [109].
In summary, transient resistance must not be neglected during P. aeruginosa infections. Different conditions, compounds and modes of growth that may take place during the infection process might transiently increase the resistance of this opportunistic pathogen to several antimicrobial treatments, thus hindering the eradication of the infecting bacterial population. Further, recent work has shown that the early appearance of tolerance mutations facilitates the evolution of AR [110], a feature of particular relevance in the case of chronic P. aeruginosa infections [111].
Infections by other Pseudomonas species with an environmental origin and biotechnological potential, as Pseudomonas putida [112,113,114,115], have also been reported, although their prevalence is much lower. Besides intrinsic resistance determinants [116,117], the acquisition of carbapenemases, as KPC-2 [113], constitutes an additional risk for the efficient treatment of infections by these pathogens.

3. Acinetobacter baumannii

Acinetobacter is another non-fermentative Gram-negative bacterial genus, firstly reported as a significant nosocomial pathogen in the late 1970s. This microorganism harbours an entire repertoire of intrinsic resistance determinants, and it easily acquired novel ARGs soon after its detection as a cause of infections [118], becoming nowadays one of the most prevalent resistant pathogens causing problems at hospitals. Unlike other pathogens discussed in the current review, and despite the Acinetobacter genus being ubiquitous, the potential primary environmental niches of Acinetobacter baumannii, the species causing most problems at hospitals, are still not well established [119]. Hence, more studies are still needed to delimitate the outside-hospitals reservoirs of A. baumannii [120]. Notably, it seems that A. baumannii presents the largest pangenome and biochemical versatility within the species of the Acinetobacter genus [121,122]. Its open pangenome contains a variety of mobile genetic elements, most notably integrons, transposons and plasmids [123], which may support the capacity of this opportunistic pathogen for acquiring ARGs. Integrons and transposons can be located in genomic islands, some of which have been dubbed islands of resistance due to the presence of multiple ARGs inside them. It has been reported that around 40% of A. baumannii pangenome is specific to each strain [121], indicating that gene exchange within this bacterial species has a certain degree of clonal specificity.
With regard to the core genome of Acinetobacter, it has been described to contain 950 families of orthologous proteins, including a large number of virulence factors [124] and at least 1590 orthologous proteins that correspond to 44% of the size of the smallest proteome of the species [121].
Due to the importance of hospital outbreaks, the population structure of A. baumannii is now well-established [125]. At least six major international clonal lineages (ICL), distributed across continents worldwide, have been described [125]. Three successful clones re-named as “international clones I-III”, among which ICLI and ICLII display MDR phenotypes that may be favouring their clonal expansion [126], are included in these lineages. A recent study, based on the analysis of almost 2500 genomes, shows that A. baumannii can be divided into two clusters. Notably, the strains of one cluster, which contain a CRISPR/Cas system, rarely harbour plasmids, indicating that CRISPR/Cas elements may modulate the acquisition of novel genes in A. baumannii [127].
Concerning human health, A. baumannii strains have been isolated primarily from hospitalized patients, and this pathogen is associated with infections of the respiratory tract, bloodstream, wound, skin and soft tissue, urinary tract and central nervous system [128]. Besides humans, A. baumannii has been isolated in veterinary medicine, infecting seriously ill animals [129], livestock and wildlife; thereby indicating that this opportunistic pathogen constitutes a One-Health problem [130].
The main limitation regarding the treatment of A. baumannii infections is the increasing prevalence of MDR isolates. Intrinsic determinants, such as the OXA-type Acinetobacter-derived cephalosporinase [131] or the RND efflux pumps AdeABC [132], AdeFGH and AdeIJK [133], stand out as major determinants of intrinsic AR in this bacterium. While the first efflux pump contributes to acquired resistance when overexpressed, the contribution of the second to this phenotype is less relevant because AdeIJK overexpression is toxic above a given threshold. AdeFGH also confers MDR when overexpressed, while some non-RND efflux systems, such as CraA, AmvA, AbeM and AbeS, have been described to be involved in A. baumannii AR too [134].
Notably, it has been shown that one-step AR mutations can be selected in vivo during the treatment of the infected patients [135]. Among them, mutations in the genes encoding the regulators of the expression of MDR efflux pumps lead to their overproduction and to associated cross-resistance to a variety of antimicrobials [136]. In addition, mutations in genes encoding outer membrane proteins, such as OmpA, CarO and OprD, also contribute to AR and modulate virulence of this opportunistic pathogen [137], providing an example of the crosstalk between virulence and AR [138].
Besides intrinsic and mutational acquired resistance, the members of this bacterial species have acquired several β-lactamases and other ARGs by HGT [139]. Among them, and in addition to the intrinsic OXA-type β-lactamase, other OXA derivatives have been found [140], frequently linked to insertion sequences (ISs) located upstream the genes encoding β-lactamases [141]. Indeed, the activity of ISs, capable of modifying the expression of genes involved in resistance when located in the right positions, seems to be also instrumental for the acquisition of the resistance phenotype [142,143]. Despite AR plasmids not being as frequent in A. baumannii as in Enterobacteriaceae [144], the plasticity of its genome [145] allows the acquisition of ARGs, many of them present in transposons and in integrons, within plasmids and the chromosome. In this regard, it is worth mentioning that more than 130 gene cassettes containing ARGs have been identified in integrons located in A. baumannii genomes [146]. Although several studies have shown the high prevalence of class 1 integrons, which often contain resistance gene cassettes, other studies carried out in Latin American countries, such as Chile, Argentina and Brazil, have also shown a wide distribution of class two integrons in this species [147,148]. It is important to notice that while extended-spectrum β-lactamases (ESBLs) as ESBLs PER-, GES- and VEB-type are the most common A. baumannii, TEM- and SHV-type ESBLs, the most prevalent in Enterobacteriaceae, are less frequently found in A. baumannii, supporting that gene exchange between these two groups of microorganisms is likely low [149], although still possible (see the example of NDM1 below).
Notably, ARGs acquired by A. baumannii are frequently clustered, forming part of genomic islands, dubbed AbaRs. These AbaRs [146] present backbones resembling Tn6019, Tn6022 and Tn6172 transposons [150] and seem to be clone-specific [151]. This feature may mean that, once an AbaR has been acquired, its mobilization to another phylogenomic A. baumannii group could be limited. Hence, HGT via plasmids or other mobile genetic elements might be on the basis of the acquisition of resistance by A. baumannii [152]. In addition, recent works indicate that ARGs-containing bacteriophages might contribute to AR spread in this microorganism [153,154]. Finally, it has been recently found that this organism can be naturally competent [155,156], opening the possibility that direct transformation could be a relevant mechanism triggering the acquisition of ARGs by this bacterium.
Currently, more studies have shown the importance of other bacterial species within the Acinetobacter genus in clinical settings [157]. Along with A. baumannii, other species such as Acinetobacter pittii and Acinetobacter nosocomialis have been frequently isolated in patients [158]. A. pittii was isolated in China, and its potential to acquire resistance to carbapenems by a mutation in blaOXA-499 has been observed [159]. Regarding A. nosocomialis, the importance of the RND-type efflux pumps, AdeIJK and AdeABC, in its resistance phenotype has been highlighted [160].
Other species of the genus have also been described in natural and clinical environments, such as Acinetobacter soli, firstly isolated from a Korean forest [161] and identified in domestic animal lice [162]. Although the first reports indicated that the microorganism came from environmental sources, it has also been found in clinical settings [163]. In China, an MDR isolate of this bacterial species containing the β-lactamase encoding genes (blaOXA-58, blaIMP-1, blaNDM-1 and blaTMB-2) caused the death of a patient under treatment [164]. Notably, it has been suggested that blaNDM-1 is a chimaera constructed in A. baumannii; a feature supporting that this species can be the origin as well as a reservoir for the transfer of this relevant carbapenemase of Enterobacteriaceae [165]. This has been further reinforced with in vitro data indicating that Acinetobacter plasmids could have contributed to the spread of blaNDM-1 in Enterobacteriaceae [166]. Also supporting this idea is the characterization of transferable plasmids containing blaNDM-1 in both A. soli and A. pittii and their mobility between the genus [167]. A. pittii isolates containing plasmids belonging to new incompatibility groups, which carry genes encoding OXA-type carbapenemases, and with the ability to transfer them to other species of the genus, have been reported too [168].
The possibility that plasmids carried by Acinetobacter spp. might be transferred to other human pathogens, hence contributing to ARGs spread, has been analysed in other studies. For instance, genomic analyses comparing Acinetobacter spp. clinical and environmental (water and soil) isolates suggested that Rep_3-type plasmids can be transferred between Acinetobacter spp. and bacteria belonging to other genera from different environments [169]. Further, the in silico analysis of 173 plasmids of a wide variety of sizes from 17 countries showed that some plasmid lineages have the capacity to replicate in many bacterial genera, while others only do it within species of the Acinetobacter genus [170]. It is important to notice that the number of different plasmid lineages harboured by A. baumannii is low, with around one-third of them containing ARGs and that gene flux among different plasmids seems to be mediated by transposons [170].
Reports on Acinetobacter spp. carrying ARGs continue to increase. Examples of them are some strains of Acinetobacter bereziniae, recently isolated from human clinical samples [171], carrying blaOXA-type [172] and MBLs encoding genes, as blaNDM-1; in both cases encoded in plasmids [173]. Another example is Acinetobacter junii strains, presenting blaNDM-1 and blaOXA-58, and isolated from hospitals [174].

4. Stenotrophomonas maltophilia

Stenotrophomonas maltophilia is a ubiquitous non-fermentative Gram-negative microorganism described in a variety of environments [175], from natural to anthropogenic niches, such as soil [176], water [177] or sediments [178,179]. Besides its role as an opportunistic pathogen, S. maltophilia is also a plant endophyte, and different strains with biotechnological value have been described [180]. This feature makes particularly relevant to distinguish between infective and non-infective S. maltophilia strains. However, there is evidence of epidemic S. maltophilia lineages [181] and it seems that, as has been described for P. aeruginosa [182,183], there are not specific clades evolving towards virulence [184,185]. Instead, the prevalence of S. maltophilia infections mainly derives from the underlying health condition of the patient, more than from specific characteristics of the isolate causing such infection. Through different genotyping methodologies, it has been described that the S. maltophilia species complex (Smc) contains multiple genospecies [178,186,187]. Four genospecies belong to the S. maltophilia sensu stricto species, which is the main cause of infections in humans and the only one that consistently expresses MBLs (Sgn1, Sgn2, Sgn3 and Sgn4) [178]. The Smc displays high genetic, ecological and phenotypic diversity [188,189] as well as heterogeneous resistance and virulence phenotypes [187,190]. In fact, this phenotypic heterogeneity mainly results from problems in species delimitation within the Smc [178,191]. This problem can be aggravated since, based on estimates of genomic mean nucleotide identity values >94%, it has been recently proposed to reclassify Stenotrophomonas africana, Pseudomonas beteli and Pseudomonas hibiscicola as S. maltophilia [188,192]; despite these species are not known to be a relevant cause of human infections.
It was in the 1980s when S. maltophilia became significantly reported as an emerging pathogen. Nowadays, it has become the third most common cause of nosocomial infections caused by non-fermentative Gram-negative bacilli. Even though it is not considered a highly virulent bacterium, it is one of the leading drug-resistant pathogens of more significant public health concern in hospitals worldwide and is associated with mortality rates between 14 and 69% in patients with bacteraemia. Although it is mainly a nosocomial pathogen, community-acquired infections are an increasing trend [193]. S. maltophilia is mostly associated with respiratory infections and acute exacerbations of COPD, followed by bloodstream infections. Less frequently, it causes infections of the skin and soft tissues, biliary and urinary tract, endocarditis, meningitis, intra-abdominal infections and endophthalmitis [194]. The most affected patients are those with previous pathologies (CF, HIV infection or cancer–particularly obstructive lung cancer-), mechanical ventilation, indwelling catheters, corticosteroid or immunosuppressant therapy, together with those hospitalized for prolonged periods or ICU admission and previous broad-spectrum antibiotics therapy [5]. In these patients, S. maltophilia infections are associated with high mortality rates [5,175,195]. Infections caused by this microorganism occur in adults and children, and the transmission to susceptible individuals takes place through direct contact with the source. Possible sources are hands of health care professionals, aerosols from CF patients, suction system tubing of dental chair units, contaminated endoscopes or tap water [175].
Treatment of infections caused by S. maltophilia is complicated given the intrinsic resistance mechanisms against most antimicrobials that this bacterium presents [196,197]. Trimethoprim/sulfamethoxazole (SXT) is currently the treatment of choice [198]; albeit, combination therapies of SXT plus ciprofloxacin, ceftazidime, tobramycin or tigecycline, which exhibit a greater activity than SXT alone, are also implemented [199,200]. However, the acquisition of resistance to SXT limits its use. Therefore, new therapeutic options are needed to tackle these infections. Ticarcillin/clavulanate or ceftazidime in combination with ciprofloxacin are the agents used in most SXT resistant infections. On the one side, ceftazidime and ticarcillin/clavulanate used to be the most effective β-lactams against S. maltophilia, but the number of resistant isolates is increasing [5]. On the other side, ciprofloxacin or newer fluoroquinolones as levofloxacin are still a helpful alternative, even though the number of resistant isolates is sizable [201]. Ultimately, recent studies have shown that minocycline [202] and colistin, alone or in combination with N-acetylcysteine [203], could be used for treating infections caused by this microorganism.
Genome sequencing of S. maltophilia clinical [195] and environmental isolates [204] indicated that several of the elements involved in the characteristic AR phenotype of this bacterial species are shared by strains isolated from different habitats. Therefore, these elements have evolved before the use of antibiotics for human therapy, as described for P. aeruginosa. In all these genomes, many genes encoding determinants of resistance to antibiotics (Table 2), such as β-lactams, cephalosporins, macrolides, fluoroquinolones, aminoglycosides or carbapenems, have been found [195,205,206]. These data show that S. maltophilia intrinsic resistance has not been acquired upon evolution in the presence of antibiotics currently used in therapy, although increased levels of resistance can be acquired by mutations or by ARGs acquisition through HGT [194,206,207].
Concerning intrinsic resistance, S. maltophilia possesses two inducible β-lactamases, L1 and L2. L1 is a broad spectrum (excluding monobactams) MBL [208], while L2 is classified as a class A clavulanic acid-sensitive cephalosporinase [209]. The expression of these enzymes, mostly controlled by the AmpR transcriptional regulator, is directly induced by the antibiotics they provide resistance to [210,211]. AmpR acts as an activator in the presence of inducers, such as β-lactams, but in the absence of them, it is a repressor of L2 expression [212].
In addition, aminoglycoside-modifying enzymes encoded in the S. maltophilia genome confer low susceptibility to several aminoglycosides [213]. Three of these enzymes have been analysed: N-aminoglycoside acetyltransferases AAC(6′)-Iz that contributes to resistance to amikacin, tobramycin, sisomicin and netilmicin [198,214]; AAC(6′)-Iak [198,213] that decreases susceptibility to several aminoglycosides, including arbekacin, kanamycin, neomycin, sisomicin or tobramycin [213]; and the aminoglycoside phosphotransferase APH(3′)-IIc [215], that confers resistance to kanamycin, neomycin, paromycin and butirosin [215].
Another mechanism of intrinsic resistance in this bacterium is the chromosomally-encoded SmQnr protein, which contributes to intrinsic resistance to quinolones [216,217,218] by protecting DNA gyrase and topoisomerases from fluoroquinolones’ activity [219].
Along with the inactivating enzymes, the major contributors to intrinsic resistance to many antimicrobial agents in S. maltophilia are the chromosome-encoded MDR efflux pumps. The best-characterized group of pumps is the RND family. Eight of these complexes (SmeABC, SmeDEF, SmeGH, SmeIJK, SmeMN, SmeOP, SmeVWX and SmeYZ) are encoded in the genome of S. maltophilia, and the role in AR of seven of them has been studied [220]. Only when the expression level is constitutively significant, as it happens with SmeYZ, SmeDEF, SmeGH, SmeIJK and SmeOP [220,221,222,223,224], do these pumps contribute to intrinsic resistance. The SmeYZ system is involved in intrinsic resistance to aminoglycosides, tetracycline, leucomycin and SXT [220]. SmeDEF overexpression is linked to quinolones, chloramphenicol, tetracycline, tigecycline, macrolides, sulfamethoxazole, trimethoprim and SXT resistance [207,225,226,227], as well as to resistance to the biocide triclosan [228,229]. SmeGH is involved in intrinsic resistance to β-lactams, quinolones, tetracycline and polymyxin B, as well as to other toxic compounds, such as menadione, tert-butyl hydroperoxide, naringenin and hexachlorophene [221]. For their part, SmeIJK and SmeOP confer resistance to aminoglycosides, tetracycline, ciprofloxacin, levofloxacin, leucomycin or minocycline [222] and nalidixic acid, doxycycline, aminoglycosides or macrolides [223], respectively.
Along with this well-characterized group, other MDR efflux pumps are encoded in the S. maltophilia genome. Among them, ATP binding cassette (ABC) efflux pumps such as, SmrA which contributes to fluoroquinolones, tetracycline and doxorubicin resistance [230]; and MacABCsm, which is involved in aminoglycosides, macrolides and polymyxins resistance [231]; and the major facilitator superfamily (MFS)-type efflux pump EmrCAB, implicated in the extrusion of nalidixic acid, erythromycin and tetrachlorosalicylanilide [232], are of clinical relevance.
This chromosomally-encoded arsenal of resistance elements, together with their low-permeability membranes, are responsible for S. maltophilia’s MDR intrinsic phenotype that is independent of the environment in which this bacterium lives [233].
In addition to their contribution to intrinsic resistance, these elements also contribute to acquired resistance when overexpressed or mutated [234]. The overexpression of either β-lactamases or MDR efflux pumps stands as the main cause of the acquisition of resistance in clinical isolates of S. maltophilia. In the case of efflux pumps, increased AR is associated with their overexpression, mostly by the acquisition of mutations in their regulators. The selection of these mutations, leading to efflux pumps’ overexpression, is particularly problematic since they lead to an MDR phenotype. For example, the most prevalent cause of S. maltophilia acquired resistance to quinolones is the overproduction of SmeDEF, mainly by mutations in the gene encoding the negative regulator SmeT [224], and of SmeVWX, by mutations in the gene encoding its SmeRv regulator [225]. Importantly, S. maltophilia is the only known bacteria in which high-level resistance to quinolones is only due to the overexpression of MDR efflux pumps, not to mutations in genes encoding quinolones targets [235,236]. Besides, evolution experiments made in the presence of tigecycline have revealed that mutations in smeT, leading to SmeDEF overexpression, constitute the first step in S. maltophilia tigecycline acquired resistance. In addition, amino acid substitutions in the gene encoding the efflux pump and, thus, changes in its structural elements are also on the basis of acquired resistance. For instance, smeH mutations are involved in the acquisition of resistance against ceftazidime, leading to cross-resistance towards other antibiotics, mainly β-lactams [237]. SmeABC, SmeIJK and SmeYZ also contribute to acquired resistance towards aminoglycosides, β-lactams and fluoroquinolones [238] when overexpressed.
Mutations in the antibiotics’ target genes are another cause of acquired resistance in this bacterium. Apart from the aforementioned smeT mutations, ribosome 30S mutations, the target of tigecycline, are among the mechanisms of tigecycline acquired resistance. Additionally, the inactivation of central carbon metabolism enzymes has also been shown to be responsible for the acquisition of AR by S. maltophilia. A group of in vitro selected mutants in which genes encoding the enzymes Eno, GmpA, GapA and Pgk were inactivated has allowed the study of mutation-driven fosfomycin resistance. This study showed that the inactivation of the Embden-Meyerhof-Parnas metabolic pathway is on the basis of this resistance [239].
Finally, not only mutations but also HGT contributes to S. maltophilia acquired resistance. Resistance to SXT may occur by the acquisition of the sul and dfrA genes present in integrons or plasmids [225]. Besides, mobile elements involved in SXT resistance, plasmid-mediated quinolone resistance genes (e.g., qnrS [240]), and β-lactamases (e.g., blaCTX-M-Gp1 [241]), have been found in S. maltophilia isolates. Despite the fact that a lysogenic phage containing the dihydrofolate reductase encoding the folA gene has been described to contribute to trimethoprim resistance in an S. maltophilia isolate [242], the role of these elements in the dissemination of ARGs in this microorganism remains to be studied in detail.
As in other species, different factors, such as medium composition, osmolarity or ionic concentrations, can induce S. maltophilia transient AR. Temperature can also modify its antibiotic susceptibility by alterations in the outer membrane LPS conformation. For instance, this bacterium is more susceptible to aminoglycosides at 37 °C than at 30 °C since the binding and/or uptake of the antibiotic is inhibited at a lower temperature [243]. The ability to form biofilms, which reduces antibiotics’ susceptibility, is a significant feature of S. maltophilia. Environmental factors, such as phosphate or chloride concentrations, temperature and aerobic or anaerobic conditions, can influence the production of biofilms, being enhanced under aerobic conditions [175]. S. maltophilia and P. aeruginosa can grow together inside dense polymicrobial biofilms in different environments, including the lungs of CF patients. This kind of growth influences their behaviour, including antibiotic susceptibility [244]. Inside these biofilms, S. maltophilia produces a diffusible signal factor that P. aeruginosa senses through the two-component sensor BptS, leading to the increased production of proteins implicated in polymyxin and colistin resistance [245].
β-lactamases and MDR efflux pumps also contribute to transient AR since their expression is inducible. The expression of β-lactamases is induced by β-lactams [210], and the MDR efflux pump’s expression increases due to the effect of different molecules. On the one side, smeDEF expression is induced by plant-derived flavonoids [246] or biocides like triclosan [229]. These molecules can bind the smeDEF repressor, SmeT, inducing the expression of this efflux pump and reducing S. maltophilia quinolone susceptibility. On the other hand, fluorescence-based analyses have uncovered smeYZ and smeVWX inducers involved in aminoglycosides and chloramphenicol or quinolone resistance, respectively. Boric acid, erythromycin, chloramphenicol and lincomycin are inducers of smeYZ [247], whereas vitamin K3 and its analogues vitamin K2 and plumbagin, as well as iodoacetate, clioquinol and sodium selenite, are smeVWX inducers [248]. Finally, the tripartite efflux pump (FuaABC), related to ABC efflux pumps, whose expression is induced by fusaric acid, contributes to transient resistance to this compound [249].

5. Burkholderia cepacia Complex

The B. cepacia complex (Bcc) is a group of closely related non-fermenting Gram-negative bacilli that comprises 22 validated species. The taxonomy of these bacteria is complex and continuously changing [250]. This complex is formed by nine genomovars, namely B. cepacia (formerly genomovar I), Burkholderia multivorans (II), Burkholderia cenocepacia (III), Burkholderia stabilis (IV), Burkholderia vietnamiensis (V), Burkholderia dolosa (VI), Burkholderia ambifaria (VII), Burkholderia anthina (VIII), Burkholderia pyrrocinia (IX) and the group or taxon K (recently split into two species: Burkholderia contaminans and Burkholderia lata) [250,251,252,253,254].
This complex has a versatile metabolism that allows it to colonize a great variety of niches [251,252,253,254]. Moreover, the complex includes species that are important opportunistic human pathogens of CF patients [252,255] or chronic granulomatosis disease, and critical nosocomial pathogens causing bacteraemia or urinary tract infections [256,257]. Although it has been stated that infections by B. cepacia could be associated with a fast lung decline and increased mortality of CF patients, dubbed the cepacia syndrome [258], recent works suggest that this statement might not always be true [259]. Further, since Bcc prevalence increases with age, lung deterioration and lung transplantation [260], these underlying conditions might also be contributing to the bad prognosis of Bcc infected patients. Although B. cenocepacia has been traditionally the most predominant cause of infections, B. multivorans is increasingly being recovered from the lungs of CF patients [261]. In addition, other members of the Bcc complex, as B. contaminans, cause infections in CF patients too [262,263]. Besides, outbreaks of healthcare-associated Bcc infections due to the contamination of pharmaceutical products have also been reported [264,265].
In addition to their relevance for human health, species of this complex are also important in agriculture because of their biocontrol and biotechnological properties. Genomovar III has been identified as a commensal of different soil types and the rhizosphere of several cultivated plants, such as maize, wheat and lupin, in natural environments [266,267]. Multilocus sequence typing (MLST) analysis of environmental and clinical isolates showed that at least 20% of the strains causing human infections are also found in nature [268]. Further, soil isolates can produce infections in both plants and animals [269]. Altogether, these findings indicate that natural ecosystems constitute a reservoir of Bcc strains with clinical relevance.
Due to its versatility and relevance in clinic and natural environments and the potential biotechnological application of some strains, the population structure of B. cenocepacia (genomovar III) has been studied in detail. It has been found that the population is in linkage disequilibrium and presents a clonal structure, with three major clones displaying variable degrees of recombination distributed worldwide [270].
One of the problems associated with Bcc infections is the low susceptibility to several antibiotics (e.g., carboxypenicillins, first and second generation cephalosporins, tetracycline or tobramycin) that this bacterial group possesses. Particularly relevant is the intrinsic resistance they all have to the last resort antibiotics polymyxins [271]. The main cause of the lack of activity of this drug against Bcc relies on the particular LPS structure of this group of microorganisms. It has been shown that the addition of 4-amino-4-deoxy-l-arabinose (Ara4N) to the lipid A component of the LPS reduces polymyxin susceptibility in different organisms [272,273]. While Ara4N synthesis is usually dispensable in different bacteria, the Ara4N biosynthetic gene cluster seems to be essential for B. cenocepacia [274]. This fact supports that the natural incorporation of Ara4N into lipid A is likely a major cause of Bcc polymyxin intrinsic resistance.
Like several other bacteria, Bcc presents in its genome genes encoding different AR determinants, including efflux pumps and inducible class A and class C β-lactamases, as PenB [275] (formerly dubbed PenA [276]) or AmpC [277], respectively, which expression is coregulated [278]. Recent work suggests that β-lactam inhibitors such as relebactam, enmetazobactam, avibactam or vaborbactam, might be useful for increasing the susceptibility to β-lactams of Bcc isolates [279].
Beyond β-lactamases, several efflux pumps are encoded in Bcc genomes. Among these systems, NorM, a member of the multidrug and toxic compound extrusion (MATE) family, has shown to play a role in polymyxin resistance, together with the aforementioned Lipid A modification [280]. Within the identified efflux pumps encoded in Bcc genomes, those present in B. cenocepacia stand out as the best studied. Namely, fourteen RND efflux pump-encoding genes have been found in the genome of B. cenocepacia [281]. These elements are able to confer resistance to clinically relevant antibiotics as aminoglycosides, chloramphenicol, fluoroquinolones and tetracyclines [281,282]. Among them, at least three are involved in intrinsic AR [283]. Notably, an MFS immunodominant efflux pump, named BcrA and involved in tetracycline and quinolones resistance, has been detected in CF patients infected with Bcc [284], suggesting that BcrA may have a relevant role in Bcc resistance to antibiotics in said patients. Besides, the finding that salicylate, a siderophore produced by B. cenocepacia, may induce the expression of an antibiotic efflux pump that confers resistance to chloramphenicol, trimethoprim and ciprofloxacin, suggests that this MDR element can be overexpressed; hence contributing to Bcc transient resistance in environments with low iron availability, such as CF patients’ lungs [285].
Besides classical AR determinants, the low susceptibility to antibiotics of Bcc is also due to global mechanisms of response to stressful compounds, such as the production of lipocalins, a family of small proteins capable of binding hydrophobic ligands. It has been shown that a soluble B. cepacia lipocalin, produced in the presence of antibiotics, allows the sequestration of such antibiotics, hence contributing to Bcc resistance [286].
Regarding genetic changes leading to acquired resistance, mutation and recombination stand as major players in this process. It has been found that B. multivorans diversifies into various clones presenting different phenotypes when causing chronic infections in CF patients; and that some loci involved in β-lactams resistance present multiple mutations in recombinogenic regions [287]. Among those, mutations in ampD, which encode a transcription factor that coregulates the expression of the two intrinsic β-lactamases AmpC and PenB, stand out among the major causes of resistance to β-lactams in Bcc. Notably, ampD is highly prone to acquire AR mutations with an estimated frequency in the range of 10−6 to 10−5 [278].
In addition to mutation-driven AR, the acquisition of ARGs through HGT by some Bcc strains has been reported. Amongst them, Type I integrons, containing the sulphonamide resistance gene sul1 and carrying the aminoglycoside resistance genes aacA4 or aacA7, or catB3, encoding a chloramphenicol acetyltransferase, are found [288]. These findings indicate that integrons may participate in the acquisition of resistance to sulfamethoxazole, chloramphenicol and aminoglycosides in Bcc.
The presence of plasmids in Bcc was studied early [289], but comprehensive information on their role in AR is still required. Something similar happens with bacteriophages. The finding of putative ARGs in prophages inserted in the chromosomes of different B. cenocepacia strains suggest that these genetic elements might be involved in the spread of resistance among Bcc [290]. However, detailed studies about the contribution of these elements in Bcc AR remain to be established.

6. Emerging Opportunistic Pathogens with Environmental Origin

Along with the opportunistic pathogens mentioned above, other environmental bacterial genera, such as Brevundimonas, Shewanella, Achromobacter, Agrobacterium, Aeromonas, Erwinia or Pantoea, among others, have been increasingly reported as responsible for emerging infectious diseases [291]. Note that while the most prevalent MDR opportunistic pathogens with a primary environmental habitat are non-fermentative Gram-negative bacteria, some environmental Enterobacteriaceae have been reported to cause human infections. Given their taxonomic relationship with highly prevalent human pathogens, as E. coli or K. pneumoniae, which easily acquire ARGs through HGT, the possibility that these environmental pathogens are a first step in the acquisition of ARGs by human bacterial pathogens [17,89,292,293] must be taken into consideration.
Brevundimonas spp. are aerobic Gram-negative bacteria that are not only isolated from soils, submarine sediments and numerous aquatic habitats; but that also cause multiple types of infections, indicating that this genus may be a more widespread pathogen than previously thought [294]. Brevundimonas diminuta and Brevundimonas vesicularis have been isolated from clinical specimens, including blood, urine and lungs of CF patients [295,296,297,298,299]. The majority of Brevundimonas infections have been found in patients with underlying diseases, and many of them are acquired in hospitals [294,300]. Importantly, Brevundimonas infections are difficult to treat, as these bacteria can be resistant to different drugs, including fluoroquinolones or β-lactams [301,302]. Resistance to fluoroquinolones may be due to mutations in gyrA, gyrB and parC [301], and resistance to β-lactams to the presence of a VIM-2 MBL [302]. In addition, tetracycline resistance genes have also been detected in environmental isolates of B. diminuta [303]. Altogether, these data indicate that these bacteria should be considered as possible causes of nosocomial infections and should be included in prevention programs. Furthermore, their suggested use in bioremediation of contaminated seas and soils [304] should be carefully re-evaluated.
Another microorganism with bioremediation potential is Shewanella algae, a marine bacterium [305] that also causes a variety of clinical symptoms in immunocompromised patients [306,307]. It has been suggested that some strains of S. algae isolated from clinical samples (skin ulcers and ear infections) [308,309,310,311] were mistakenly identified as Shewanella putrefaciens [312,313,314], a very close bacterial species [313]. A recent study has described the presence of β-lactams resistant clones of S. algae along the Italian Adriatic coast, containing AmpC and OXA-55-like β-lactamases [315]. Further, these authors have described the possible role of S. algae as a reservoir of ARGs, such as qnrA and β-lactamase genes (that confer resistance to quinolones and β-lactams, respectively), which could be transferred from the aquatic microbiota of Italian fish farms to bacteria of medical interest [316]. Actually, it has been proposed that S. algae are the origin of the quinolone resistance gene qnrA, widely distributed among plasmids present in several organisms [317], and different Shewanella species (as well as A. baumannii, see above) are considered as potential origins of some OXA-type β-lactamases [318]. Besides their contribution as progenitors of mobile ARGs, Shewanella can be involved in such mobility too. In fact, a plasmid harbouring several ARGs has been recently identified in Shewanella xiamenensis [319]. Although serious infections caused by Shewanella have been described [320,321], the rarity of these infections means that treatment guidelines have not been defined yet.
The genus Achromobacter is found in soils and aquatic environments, although some isolates can colonize the human intestinal tract, becoming opportunistic pathogens in immunosuppressed patients [322]. These bacteria can cause bacteraemia, meningitis and urinary tract infections [323,324,325]. Moreover, Achromobacter genus-belonging bacteria have also been isolated from CF patients [326,327,328]. Achromobacter xylosoxidans is the predominantly reported species among CF clinical isolates [322], but other Achromobacter species have also been isolated from these patients, such as Achromobacter ruhlandiiand [329]. They are intrinsically resistant to several drugs [328] due to the presence in their genomes of genes encoding RND MDR efflux pumps that extrude cephalosporins, aztreonam, carbapenems, quinolones, chloramphenicol, tetracyclines and erythromycin [330,331], as well as to the activity of β-lactamases [332,333]. Moreover, they are becoming increasingly resistant to carbapenems [328]. Furthermore, a recent study has described patient-to-patient transmission and AR development in different Achromobacter species [334]. Therefore, these species should be included in preventive programs.
The genus Agrobacterium is a recognized group of soil and plant-pathogenic bacteria that has also been implicated in human opportunistic infections, particularly Agrobacterium radiobacter (also known as Rhizobium radiobacter). Infections caused by these bacteria include bacteraemia, peritonitis and urinary tract infections, and they have been frequently associated with the use of intravascular devices in immunocompromised patients [335,336,337]. Further, A. radiobacter has been recently described to cause ocular infections, and it was identified in polymicrobial keratitis cases [338]. Although there is not much information about the intrinsic AR of these bacteria, the presence of RND efflux pumps in the genus Agrobacterium [339,340] suggests that these bacteria may present low susceptibility to different drugs. Further, the finding of an A. radiobacter clinical isolates carrying different antibiotic-inactivating enzymes [341] indicate that this microorganism may possess a wide set of AR determinants.
Aeromonas are Gram-negative bacteria with an aquatic environmental primary habitat that have also been suggested to behave as opportunistic pathogens [342]. Although there are controversial data about the role of these bacteria in human pathogenesis [343,344,345], different studies have described a significant correlation between diseases and the production of different virulence factors, such as haemolysins and enterotoxins [346,347]. The principal sources of these infections are contaminated water and foods, mainly inadequately cooked seafood and oysters [348,349]. In particular, Aeromonas intestinalis, Aeromonas enterica, Aeromonas crassostreae and Aeromonas aquatilis have been recently identified as representative species of Aeromonas with pathogenicity for both humans and aquatic organisms [350,351]. Aeromonas spp. are difficult to treat due to their intrinsic resistance to β-lactams, which results from a high constitutive expression of the gene encoding the β-lactamase AmpC, the low permeability of their external membrane and the activity of several outer membrane proteins [351,352]. In addition, Aeromonas species can also acquire resistance to β-lactams, such as ampicillin, and drugs from other structural families, such as erythromycin, tetracycline or chloramphenicol, by the acquisition of ARGs [353,354]. Moreover, these bacteria may have importance in aquatic environments as reservoirs of ARGs [355,356]. In this regard, it is worth mentioning that the analysis of the bacterial lineages likely associated with the dissemination of ARGs in a wastewater treatment plant indicate that Aeromonas could be a hub for such dissemination [11].
Erwinia is a genus of Enterobacteriales ubiquitous in the environment, especially in aquatic ecosystems and soils [357]. It mainly comprises phytopathogenic species, such as Erwinia amylovora, the first pathogen shown to cause disease in plants (i.e., the fire blight); Erwinia persinicus, which infects a wide range of hosts (e.g., tomatoes, cucumbers and bean pods); or Erwinia carotovora, among others. Strikingly, these phytopathogens and other plant-associated non-pathogenic Erwinia species (i.e., Erwinia billingiae and Erwinia tasmaniensis) have been occasionally found infecting animals, including humans [358]. For instance, E. carotovora and E. persinicus have exhibited pathogenicity against invertebrate infection models [357,359], and the latter has also been isolated from a human urinary tract infection [360]; whereas, E. billingiae and other non-phytopathogenic Erwinia strains can cause cutaneous infections, septic arthritis, brain abscesses or bacteraemia in humans [361,362,363,364]. Since these examples are quite unusual, the AR determinants that these species could harbour have not been sufficiently studied. However, there are reports about ARGs present in E. amylovora, like strAB, which codes for a phosphotransferase that confers resistance to streptomycin, and that has been likely acquired by non-pathogenic epiphytic bacteria also present in plant hosts [365]. In addition, some E. amylovora strains resistant to oxolinic acid, most likely mediated by chromosomal mutations, have been described [366]. Considering all this information, the potential of the Erwinia genus to become an opportunistic human pathogen, as other bacterial species described here, should be closely monitored.
Another Gram-negative genus within the Enterobacteriaceae family is Pantoea, which includes 20 species isolated from different aquatic and terrestrial environments. Although many Pantoea isolates are misidentified, they have been described in association with plants and animals; mainly insects, but also birds, fish, bears, ruminants and importantly, humans [367,368,369]. The ability of this bacterial group to compete and survive in different niches has made it attractive for biotechnological uses. Water and soil isolates have been used for industrial applications, as bioremediation, since they are able to degrade many products; or agricultural purposes because they compete with plant pathogens and induce plant defences [370,371]. Besides being a plant pathogen, Pantoea has been recently identified in nosocomial environments. Different Pantoea species have been isolated from both immunocompetent and immunocompromised patients from wounds, blood, skin, stool, cysts and abscesses, as well as from urethra, trachea and oropharyngeal swabs [368]. Opportunistic infections in humans caused by Pantoea include septicaemia, pneumonia, septic arthritis, wound infections and meningitis [372]. As it happens with other pathogens with an environmental origin, clinical and environmental isolates are phylogenetically indistinguishable. Even more, Pantoea species considered primarily plant pathogens can be isolated from humans [367,369]. The most prevalent species infecting humans are Pantoea agglomerans and Pantoea septica [369]. Other clinically-relevant species include Pantoea dispersa, causing bacteraemia and neonatal sepsis [373], Pantoea brenneri and Pantoea conspicua, isolated from human sputum and blood, respectively [374,375]. Besides, clinical reports demonstrated cases of pneumonia and death in children with comorbidities where the causative agent was identified as an MDR P. agglomerans, resistant to third-generation cephalosporins, carbapenems, aminoglycosides and ciprofloxacin [376]. However, Pantoea’s AR determinants are mostly unexplored. Recently, a study has found that a foodborne P. agglomerans isolate possesses RND, ABC and MFS antibiotic efflux pumps such as MdtABC, MsbA or EmrAB, and antibiotic target modifiers that provide resistance to antibiotics such as macrolides, fluoroquinolones, tetracyclines or aminoglycosides [372]. Accordingly, further studies are needed to validate the ARGs of this opportunistic pathogen.
Altogether, these data indicate that natural environments are an important primary source of opportunistic pathogens. Since humans, animals and natural environments are interconnected, One-Health approaches [8] are required to limit the spread and evolution of AR.

7. Ecological Role of Antibiotic Resistance Determinants Outside Clinical Settings

The environmental origin of different opportunistic bacteria indicates that the mechanisms of virulence and AR with a current role in human infection present a different and unique function in the natural environments where these bacteria emerge. Indeed, the fact that intrinsic ARGs may have other functional roles besides AR has been previously discussed [17,377,378,379,380]. While some of these functions deal with basic aspects of bacterial physiology, such as peptidoglycan recycling [381], some others are related to bacterial interactions with other elements of the biosphere and hence, have ecological value. This includes not only ARGs but also situations that trigger transient AR. For instance, it has been demonstrated that increased production of alginate, a key element for P. aeruginosa biofilm formation, protects this bacterial species against its protozoan predators in nature [382]. Concerning ARGs, bacterial MDR efflux pumps stand as relevant elements modulating bacterial interactions with the environment. These ARGs are ancient elements that extrude not only antibiotics but also a wide range of non-antibiotic substrates. Further, the facts that efflux pumps are conserved within a species and between species [383,384], that their expression may be induced by host-produced compounds [246,385,386,387,388,389,390,391,392] (such as bile salts or fatty acids, plant-produced compounds or QS signals, from humans, plants and bacteria, respectively), and that these systems are able to extrude non-antibiotic substrates [393] (such as QS signals, bacterial metabolites, or plant-produced compounds [390,392,394,395,396,397,398,399]), indicates that they play important roles in the adaptation of bacterial physiology to changing environments. In this review, we discuss the role of efflux pumps outside clinical settings, focusing on bacterial interactions in the rhizosphere.
The rhizosphere is a natural ecosystem that comprises the plant roots and microbial community present in the surrounding soil. Within this ecosystem, soil bacteria and plants affect each other, leading to a feedback system that drives the ecology and evolution of both organisms [400]. Accordingly, the evolution of the microbial community is the result of either the trade-offs associated with overcoming the plants’ defence or the specific benefits associated with the host plant colonization. In this sense, plants’ roots, apart from providing mechanical support and allowing the absorption of water and nutrients by plants, exudate a wide array of natural products into the rhizosphere [401]. This extrusion modifies soil composition and provides both nutrients for bacterial growth and defensive secondary metabolites. Therefore, roots shape the composition and dynamics of microbial communities, as only bacteria capable of dealing with root exudates are present in the rhizosphere, but they also drive the evolution of plant pathogens [402]. The selection of more virulent mutants that can evade plant defences [403,404] and of mutants that present an improved capacity to metabolize plant-produced nutrients is the driving force of this evolution [405]. Even more, microorganisms present different mechanisms that allow them to deal with root exudates, such as the flavonoid-responsive family of RND efflux pumps. These mechanisms of resistance have been identified in different plant-associated bacteria such as Agrobacterium tumefaciens [340], Pseudomonas syringae [390,406], E. amylovora [392,407], Bradyrhizobium japonicum [408], Xanthomonas axonopodis, Ralstonia solanacearum [409], S. maltophilia [246] and Sinorhizobium meliloti [410].
As mentioned above, MexAB-OprM is an important MDR determinant of the human opportunistic pathogen P. aeruginosa [411,412], which contributes to its intrinsic resistance to several antibiotics (quinolones, macrolides, tetracycline, chloramphenicol and β-lactams) [413]. Besides, this pump is a relevant mechanism for acquiring AR in clinical settings [414] since mexAB-oprM overexpressing mutants are selected in the infected patients [415]. MexAB-OprM is also able to extrude monoterpenes and related alcohols present in the tea tree (Melaleuca alternifolia) [394], indicating a role in natural environments that was probably acquired before that of antibiotic resistance at clinical settings. In fact, plant flavonoids induce the expression of mexAB-oprM in P. syringae, the causal agent of bacterial speck in tomato plants, which allows colonization of these plants [390]. These compounds are inhibitors of motility and the type III secretion system in P. syringae via the GacS/GacA two-component system [416,417]. Therefore, one of the roles of MexAB-OprM in natural environments is extruding flavonoids to avoid the inhibition of virulence and hence, allowing the colonization of tomato plants. In fact, flavonoids also regulate the capacity of other plant-associated bacteria to colonize plants, such as E. amylovora, A. tumefaciens, X. axonopodis and S. maltophilia. These effectors are inducers and may also be substrates of efflux pumps [246,407,418,419]. These Red-Queen adaptive coevolution phenomena indicate that the original role of bacterial efflux pumps may be the extrusion of plant-derived anti-virulence compounds, among others. Therefore, the screening of natural or natural-like compounds that act as both inducers and substrates of efflux pumps of clinical relevance could serve to identify virulence inhibitors that could be potentially combined with antibiotics in new therapeutic strategies to control bacterial infections caused by environmental pathogens as P. aeruginosa [420].
Root exudates not only avoid colonization by pathogenic bacteria but also recruit nitrogen-fixing and growth-promoting bacteria [421]. Many plant species, mainly legumes, present an intimate association with nitrogen-fixing bacteria and, again, the above-mentioned flavonoids are involved in establishing these associations [422]. This has been observed in S. meliloti [410,423] and B. japonicum [408], in which flavonoids are also inducers of efflux pumps. Additionally, roots also attract bacteria able to promote plant growth by the extrusion of carbohydrates, amino acids and benzoxazinoids [424,425]. Once again, efflux pumps may be mediating these associations.
Efflux pumps also play essential roles in bacteria-bacteria interactions within the host plant, where there is competition for space and nutrients. Cell-cell interactions are controlled by the QS system, which allows cooperation within a species to colonize a given environment and inter-species communication. In this regard, it is known that the AR determinants MexAB-OprM and MexCD-OprJ of P. aeruginosa [411,412,426] modulate QS-responses and host-pathogen interactions, either by the extrusion [427,428,429,430] or by the impaired production [431] of QS signals or their metabolic precursors. While bacteria from the rhizosphere produce QS signals to coordinate plant colonization [432], plants may secrete compounds similar to bacterial N-acyl-homoserine lactones (AHLs) through root exudation [433,434], something that is known as Quorum Quenching. For instance, the red seaweed Delisea pulchra produces halogenated furanones that interfere with the AHL regulatory system of several Gram-negative bacteria [435,436]. In addition, it is known that certain bacteria also possess the ability to quench QS by enzymatic degradation of AHL signals [437]. This is the case of a Bacillus acyl-homoserine lactonase enzyme able to hydrolyse the lactone bond of AHL compounds of the plant pathogen E. carotovora [438].
Finally, another relevant role of efflux pumps in bacteria-bacteria interactions within the plant host is the extrusion of antimicrobial compounds produced by other bacterial species. For example, E. amylovora and P. agglomerans (a biocontrol agent for fire blight) co-colonize rosaceous plants [439,440], but the last one impedes colonization of stigmas of apple and pear plants by E. amylovora by effectively inhibiting its growth [441]. However, this microorganism can reach high-density populations when the expression of norM is induced (at 18 °C [439]), indicating that this efflux pump extrudes antimicrobial compounds produced by P. agglomerans [396].
All in all, these data indicate that bacterial efflux pumps are much more than AR determinants. They are relevant elements for the physiology of microorganisms in natural ecosystems. In this sense, it is important to keep in mind that evolution is similar to a tinkerer [442], which produces new functions from old materials and not from scratch.
Table 1. Clinically relevant MDR efflux pumps in P. aeruginosa
Table 1. Clinically relevant MDR efflux pumps in P. aeruginosa
Efflux PumpMain RegulatorsSubstrate RangeResistanceReferences
MexAB-OprMMexR, NalD, NalCβ-lactams (excepting imipenem), quinolones, macrolides, tetracyclines, chloramphenicolIR *, AR **,
TR ***
[384]
MexCD-OprJNfxBPenicillin, cefepime, cefpirome, meropenem, quinolones, macrolides, tetracyclines, chloramphenicolAR, TR[413]
MexEF-OprNMexT, MexSCarbapenems, quinolones, chloramphenicolAR, TR[443]
MexXY-OprMMexZPenicillin, cefepime, cefpirome, meropenem, quinolones, macrolides, tetracyclines, chloramphenicol, aminoglycosidesIR, AR, TR[413]
* Intrinsic (IR) ** Acquired (AR) and *** Transient (TR) antibiotic resistance.
Table 2. Main antibiotic resistance determinants encoded in S. maltophilia genome.
Table 2. Main antibiotic resistance determinants encoded in S. maltophilia genome.
GeneProductDrug ResistanceType of ResistanceReferences
L1Class B3 Zn2+-dependent MBLβ-lactams (except monobactams)IR *, TR ***[208,211]
L2Class A clavulanic acid-sensitive cephalosporinaseβ-lactamsIR, TR[209,211]
aac(6′)-IzN-Aminoglycoside acetyltransferaseAmikacin, tobramycin, sisomicin, netilmicinIR[214]
aac(6′)-IakN-Aminoglycoside acetyltransferaseArbekacin, kanamycin, neomycin, sisomicin, tobramycinIR[213]
aph(3′)-IIcAminoglycoside phosphotransferaseKanamycin, neomycin, paromycin, butirosinIR[215]
SmqnrPentapeptide repeat proteinQuinolonesIR, AR **[218,219,240]
smeYZRND efflux pumpAminoglycosides, tetracycline, leucomycin, SXTIR, AR, TR[247,444]
smeDEFRND efflux pumpFluoroquinolones, chloramphenicol tetracycline, tigecycline, macrolides, sulfamethoxazole, trimethoprim, SXTIR, AR, TR[207,225,229,445,446]
smeGHRND efflux pumpβ-lactams, fluoroquinolones, tetracycline, polymyxin B, ceftazidimeIR, AR[221]
smeIJKRND efflux pumpAminoglycosides, tetracycline, ciprofloxacin, levofloxacin, leucomycin, minocyclineIR, AR[222]
smeOPRND efflux pumpNalidixic acid, doxycycline, aminoglycosides, macrolidesIR[223]
smeVWXRND efflux pumpQuinolones, chloramphenicol, trimethoprim/sulfamethoxazoleAR, TR[225,226,247,248]
smeABCRND efflux pumpAminoglycosides, β-lactams and fluoroquinolonesAR[238]
smrAABC efflux pumpFluoroquinolones, tetracycline, doxorubicinND[230]
macABCsmABC efflux pumpAminoglycosides, macrolides, polymyxinsIR[231]
emrCABsmMFS efflux pumpNalidixic acid, erythromycin, CCCP, tetrachlorosalicylanilideIR[232]
fuaABCFusaric acid tripartite efflux pumpFusaric acidTR[249]
* Intrinsic (IR) ** Acquired (AR) and *** Transient (TR) antibiotic resistance.

Author Contributions

All authors contributed to the design of the structure and the writing of the review. All authors have read and agreed to the published version of the manuscript.

Funding

Work in the laboratory is supported by Instituto de Salud Carlos III (grant RD16/0016/0011)—co-financed by the European Development Regional Fund “A Way to Achieve Europe”, by grant S2017/BMD-3691 InGEMICS-CM, funded by Comunidad de Madrid (Spain) and European Structural and Investment Funds and by the Spanish Ministry of Economy and Competitivity (BIO2017-83128-R). PL is the recipient of a FPU fellowship and TGG of a FPI fellowship, both from MINECO. LEOS is supported by a postdoctoral fellowship from Consejo Nacional de Ciencia y Tecnología (CONACyT-México). The funders had no role in study design, or the decision to submit the work for publication.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Martínez, J.L.; Baquero, F.; Nombela, C.; Cassell, G.H.; Gutiérrez-Fuentes, J.A. Evolution of bacterial opportunistic pathogens. Evol. Biol. Bact. Fungal Pathog. 2008, 9, 85–91. [Google Scholar]
  2. Waksman, S.A.; Woodruff, H.B. The soil as a source of microorganisms antagonistic to disease-producing bacteria. J. Bacteriol. 1940, 40, 581–600. [Google Scholar] [CrossRef] [Green Version]
  3. Enoch, D.A.; Birkett, C.I.; Ludlam, H.A. Non-fermentative Gram-negative bacteria. Int. J. Antimicrob. Agents 2007, 29 (Suppl. S3), S33–S41. [Google Scholar] [CrossRef]
  4. Brown, S.P.; Cornforth, D.M.; Mideo, N. Evolution of virulence in opportunistic pathogens: Generalism, plasticity, and control. Trends Microbiol. 2012, 20, 336–342. [Google Scholar] [CrossRef] [Green Version]
  5. Chang, Y.T.; Lin, C.Y.; Chen, Y.H.; Hsueh, P.R. Update on infections caused by Stenotrophomonas maltophilia with particular attention to resistance mechanisms and therapeutic options. Front. Microbiol. 2015, 6, 893. [Google Scholar] [CrossRef] [PubMed]
  6. Berendonk, T.U.; Manaia, C.M.; Merlin, C.; Fatta-Kassinos, D.; Cytryn, E.; Walsh, F.; Burgmann, H.; Sorum, H.; Norstrom, M.; Pons, M.N.; et al. Tackling antibiotic resistance: The environmental framework. Nat. Rev. Microbiol. 2015, 13, 310–317. [Google Scholar] [CrossRef] [PubMed]
  7. Baquero, F.; Martinez, J.L.; Canton, R. Antibiotics and antibiotic resistance in water environments. Curr. Opin. Biotechnol. 2008, 19, 260–265. [Google Scholar] [CrossRef]
  8. Hernando-Amado, S.; Coque, T.M.; Baquero, F.; Martinez, J.L. Defining and combating antibiotic resistance from One Health and Global Health perspectives. Nat. Microbiol. 2019, 4, 1432–1442. [Google Scholar] [CrossRef]
  9. Martinez, J.L. Natural antibiotic resistance and contamination by antibiotic resistance determinants: The two ages in the evolution of resistance to antimicrobials. Front. Microbiol. 2012, 3, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Karkman, A.; Parnanen, K.; Larsson, D.G.J. Fecal pollution can explain antibiotic resistance gene abundances in anthropogenically impacted environments. Nat. Commun. 2019, 10, 80. [Google Scholar] [CrossRef]
  11. Narciso-da-Rocha, C.; Rocha, J.; Vaz-Moreira, I.; Lira, F.; Tamames, J.; Henriques, I.; Martinez, J.L.; Manaia, C.M. Bacterial lineages putatively associated with the dissemination of antibiotic resistance genes in a full-scale urban wastewater treatment plant. Environ. Int. 2018, 118, 179–188. [Google Scholar] [CrossRef] [PubMed]
  12. Pärnänen, K.M.M.; Narciso-da-Rocha, C.; Kneis, D.; Berendonk, T.U.; Cacace, D.; Do, T.T.; Elpers, C.; Fatta-Kassinos, D.; Henriques, I.; Jaeger, T.; et al. Antibiotic resistance in European wastewater treatment plants mirrors the pattern of clinical antibiotic resistance prevalence. Sci. Adv. 2019, 5, eaau9124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hendriksen, R.S.; Munk, P.; Njage, P.; van Bunnik, B.; McNally, L.; Lukjancenko, O.; Roder, T.; Nieuwenhuijse, D.; Pedersen, S.K.; Kjeldgaard, J.; et al. Global monitoring of antimicrobial resistance based on metagenomics analyses of urban sewage. Nat. Commun. 2019, 10, 1124. [Google Scholar] [CrossRef] [PubMed]
  14. Lira, F.; Vaz-Moreira, I.; Tamames, J.; Manaia, C.M.; Martínez, J.L. Metagenomic analysis of an urban resistome before and after wastewater treatment. Sci. Rep. 2020, 10, 8174. [Google Scholar] [CrossRef]
  15. Larsson, D.G.J.; Andremont, A.; Bengtsson-Palme, J.; Brandt, K.K.; de Roda Husman, A.M.; Fagerstedt, P.; Fick, J.; Flach, C.F.; Gaze, W.H.; Kuroda, M.; et al. Critical knowledge gaps and research needs related to the environmental dimensions of antibiotic resistance. Environ. Int. 2018, 117, 132–138. [Google Scholar] [CrossRef]
  16. Martinez, J.L. Bacterial pathogens: From natural ecosystems to human hosts. Environ. Microbiol. 2013, 15, 325–333. [Google Scholar] [CrossRef] [PubMed]
  17. Martinez, J.L. The role of natural environments in the evolution of resistance traits in pathogenic bacteria. Proc. Biol. Sci. 2009, 276, 2521–2530. [Google Scholar] [CrossRef] [Green Version]
  18. Silby, M.W.; Winstanley, C.; Godfrey, S.A.; Levy, S.B.; Jackson, R.W. Pseudomonas genomes: Diverse and adaptable. FEMS Microbiol. Rev. 2011, 35, 652–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Green, S.K.; Schroth, M.N.; Cho, J.J.; Kominos, S.K.; Vitanza-jack, V.B. Agricultural plants and soil as a reservoir for Pseudomonas aeruginosa. Appl. Microbiol. 1974, 28, 987–991. [Google Scholar] [CrossRef]
  20. Myers, G.E. Survival of pathogenic bacteria in crude oil. J. Environ. Pathol. Toxicol. Oncol. 1984, 5, 79–87. [Google Scholar]
  21. Howard, I.; Espigares, E.; Lardelli, P.; Martin, J.L.; Espigares, M. Evaluation of microbiological and physicochemical indicators for wastewater treatment. Environ. Toxicol. 2004, 19, 241–249. [Google Scholar] [CrossRef]
  22. Nonaka, L.; Inubushi, A.; Shinomiya, H.; Murase, M.; Suzuki, S. Differences of genetic diversity and antibiotics susceptibility of Pseudomonas aeruginosa isolated from hospital, river and coastal seawater. Environ. Microbiol. Rep. 2010, 2, 465–472. [Google Scholar] [CrossRef]
  23. Crone, S.; Vives-Florez, M.; Kvich, L.; Saunders, A.M.; Malone, M.; Nicolaisen, M.H.; Martinez-Garcia, E.; Rojas-Acosta, C.; Catalina Gomez-Puerto, M.; Calum, H.; et al. The environmental occurrence of Pseudomonas aeruginosa. Apmis 2020, 128, 220–231. [Google Scholar] [CrossRef]
  24. Walker, T.S.; Bais, H.P.; Deziel, E.; Schweizer, H.P.; Rahme, L.G.; Fall, R.; Vivanco, J.M. Pseudomonas aeruginosa—Plant root interactions. Pathogenicity, biofilm formation, and root exudation. Plant Physiol. 2004, 134, 320–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Miyata, S.; Casey, M.; Frank, D.W.; Ausubel, F.M.; Drenkard, E. Use of the Galleria mellonella caterpillar as a model host to study the role of the type III secretion system in Pseudomonas aeruginosa pathogenesis. Infect. Immun. 2003, 71, 2404–2413. [Google Scholar] [CrossRef] [Green Version]
  26. Ruiz-Diez, B.; Sanchez, P.; Baquero, F.; Martinez, J.L.; Navas, A. Differential interactions within the Caenorhabditis elegans-Pseudomonas aeruginosa pathogenesis model. J. Theor. Biol. 2003, 225, 469–476. [Google Scholar] [CrossRef]
  27. Clatworthy, A.E.; Lee, J.S.; Leibman, M.; Kostun, Z.; Davidson, A.J.; Hung, D.T. Pseudomonas aeruginosa infection of zebrafish involves both host and pathogen determinants. Infect. Immun. 2009, 77, 1293–1303. [Google Scholar] [CrossRef] [Green Version]
  28. Snouwaert, J.N.; Brigman, K.K.; Latour, A.M.; Malouf, N.N.; Boucher, R.C.; Smithies, O.; Koller, B.H. An animal model for cystic fibrosis made by gene targeting. Science 1992, 257, 1083–1088. [Google Scholar] [CrossRef] [Green Version]
  29. Obritsch, M.D.; Fish, D.N.; MacLaren, R.; Jung, R. Nosocomial infections due to multidrug-resistant Pseudomonas aeruginosa: Epidemiology and treatment options. Pharmacotherapy 2005, 25, 1353–1364. [Google Scholar] [CrossRef]
  30. Carilla-Latorre, S.; Calvo-Garrido, J.; Bloomfield, G.; Skelton, J.; Kay, R.R.; Ivens, A.; Martinez, J.L.; Escalante, R. Dictyostelium transcriptional responses to Pseudomonas aeruginosa: Common and specific effects from PAO1 and PA14 strains. BMC Microbiol. 2008, 8, 109. [Google Scholar] [CrossRef] [Green Version]
  31. Pirnay, J.P.; Bilocq, F.; Pot, B.; Cornelis, P.; Zizi, M.; Van Eldere, J.; Deschaght, P.; Vaneechoutte, M.; Jennes, S.; Pitt, T.; et al. Pseudomonas aeruginosa population structure revisited. PLoS ONE 2009, 4, e7740. [Google Scholar] [CrossRef] [Green Version]
  32. Bassetti, M.; Vena, A.; Croxatto, A.; Righi, E.; Guery, B. How to manage Pseudomonas aeruginosa infections. Drugs Context 2018, 7, 212527. [Google Scholar] [CrossRef]
  33. Martinez-Solano, L.; Macia, M.D.; Fajardo, A.; Oliver, A.; Martinez, J.L. Chronic Pseudomonas aeruginosa infection in chronic obstructive pulmonary disease. Clin. Infect. Dis. 2008, 47, 1526–1533. [Google Scholar] [CrossRef] [Green Version]
  34. Lambiase, A.; Raia, V.; Del Pezzo, M.; Sepe, A.; Carnovale, V.; Rossano, F. Microbiology of airway disease in a cohort of patients with cystic fibrosis. BMC Infect. Dis. 2006, 6, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kc, R.; Adhikari, S.; Bastola, A.; Devkota, L.; Bhandari, P.; Ghimire, P.; Adhikari, B.; Rijal, K.R.; Banjara, M.R.; Ghimire, P. Opportunistic Respiratory Infections in HIV Patients Attending Sukraraj Tropical and Infectious Diseases Hospital in Kathmandu, Nepal. HIV/AIDS 2019, 11, 357–367. [Google Scholar] [CrossRef] [Green Version]
  36. Samonis, G.; Vardakas, K.Z.; Kofteridis, D.P.; Dimopoulou, D.; Andrianaki, A.M.; Chatzinikolaou, I.; Katsanevaki, E.; Maraki, S.; Falagas, M.E. Characteristics, risk factors and outcomes of adult cancer patients with extensively drug-resistant Pseudomonas aeruginosa infections. Infection 2014, 42, 721–728. [Google Scholar] [CrossRef]
  37. Turner, K.H.; Everett, J.; Trivedi, U.; Rumbaugh, K.P.; Whiteley, M. Requirements for Pseudomonas aeruginosa acute burn and chronic surgical wound infection. PLoS Genet. 2014, 10, e1004518. [Google Scholar] [CrossRef] [Green Version]
  38. Cole, S.J.; Records, A.R.; Orr, M.W.; Linden, S.B.; Lee, V.T. Catheter-associated urinary tract infection by Pseudomonas aeruginosa is mediated by exopolysaccharide-independent biofilms. Infect. Immun. 2014, 82, 2048–2058. [Google Scholar] [CrossRef] [Green Version]
  39. Gbejuade, H.O.; Lovering, A.M.; Webb, J.C. The role of microbial biofilms in prosthetic joint infections. Acta Orthop. 2015, 86, 147–158. [Google Scholar] [CrossRef]
  40. Hoiby, N.; Ciofu, O.; Bjarnsholt, T. Pseudomonas aeruginosa biofilms in cystic fibrosis. Future Microbiol. 2010, 5, 1663–1674. [Google Scholar] [CrossRef]
  41. Smith, R.S.; Iglewski, B.H.P. aeruginosa quorum-sensing systems and virulence. Curr. Opin. Microbiol. 2003, 6, 56–60. [Google Scholar] [CrossRef]
  42. Olivares, J.; Bernardini, A.; Garcia-Leon, G.; Corona, F.; Sanchez, M.B.; Martinez, J.L. The intrinsic resistome of bacterial pathogens. Front. Microbiol. 2013, 4, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Santajit, S.; Indrawattana, N. Mechanisms of Antimicrobial Resistance in ESKAPE Pathogens. BioMed Res. Int. 2016, 2016, 2475067. [Google Scholar] [CrossRef] [Green Version]
  44. Rello, J.; Kalwaje Eshwara, V.; Lagunes, L.; Alves, J.; Wunderink, R.G.; Conway-Morris, A.; Rojas, J.N.; Alp, E.; Zhang, Z. A global priority list of the TOp TEn resistant Microorganisms (TOTEM) study at intensive care: A prioritization exercise based on multi-criteria decision analysis. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2019, 38, 319–323. [Google Scholar] [CrossRef]
  45. Gaspar, M.C.; Couet, W.; Olivier, J.C.; Pais, A.A.; Sousa, J.J. Pseudomonas aeruginosa infection in cystic fibrosis lung disease and new perspectives of treatment: A review. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2013, 32, 1231–1252. [Google Scholar] [CrossRef]
  46. Fraile-Ribot, P.A.; Zamorano, L.; Orellana, R.; Del Barrio-Tofino, E.; Sanchez-Diener, I.; Cortes-Lara, S.; Lopez-Causape, C.; Cabot, G.; Bou, G.; Martinez-Martinez, L.; et al. Activity of Imipenem-Relebactam against a Large Collection of Pseudomonas aeruginosa Clinical Isolates and Isogenic beta-Lactam-Resistant Mutants. Antimicrob. Agents Chemother. 2020, 64, e02165-19. [Google Scholar] [CrossRef]
  47. Pang, Z.; Raudonis, R.; Glick, B.R.; Lin, T.J.; Cheng, Z. Antibiotic resistance in Pseudomonas aeruginosa: Mechanisms and alternative therapeutic strategies. Biotechnol. Adv. 2019, 37, 177–192. [Google Scholar] [CrossRef] [PubMed]
  48. Blanco, P.; Sanz-Garcia, F.; Hernando-Amado, S.; Martinez, J.L.; Alcalde-Rico, M. The development of efflux pump inhibitors to treat Gram-negative infections. Expert Opin. Drug Discov. 2018, 13, 919–931. [Google Scholar] [CrossRef]
  49. Imperi, F.; Fiscarelli, E.V.; Visaggio, D.; Leoni, L.; Visca, P. Activity and Impact on Resistance Development of Two Antivirulence Fluoropyrimidine Drugs in Pseudomonas aeruginosa. Front. Cell. Infect. Microbiol. 2019, 9, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Ferrer-Espada, R.; Sanchez-Gomez, S.; Pitts, B.; Stewart, P.S.; Martinez-de-Tejada, G. Permeability enhancers sensitize beta-lactamase-expressing Enterobacteriaceae and Pseudomonas aeruginosa to beta-lactamase inhibitors, thereby restoring their beta-lactam susceptibility. Int. J. Antimicrob. Agents 2020, 56, 105986. [Google Scholar] [CrossRef]
  51. Roemhild, R.; Gokhale, C.S.; Dirksen, P.; Blake, C.; Rosenstiel, P.; Traulsen, A.; Andersson, D.I.; Schulenburg, H. Cellular hysteresis as a principle to maximize the efficacy of antibiotic therapy. Proc. Natl. Acad. Sci. USA 2018, 115, 9767–9772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Hernando-Amado, S.; Sanz-García, F.; Martínez, J.L. Rapid and robust evolution of collateral sensitivity in Pseudomonas aeruginosa antibiotic-resistant mutants. Sci. Adv. 2020, 6, eaba5493. [Google Scholar] [CrossRef] [PubMed]
  53. Barbosa, C.; Mahrt, N.; Bunk, J.; Grasser, M.; Rosenstiel, P.; Jansen, G.; Schulenburg, H. The Genomic Basis of Rapid Adaptation to Antibiotic Combination Therapy in Pseudomonas aeruginosa. Mol. Biol. Evol. 2021, 38, 449–464. [Google Scholar] [CrossRef]
  54. Laborda, P.; Martínez, J.L.; Hernando-Amado, S. Convergent phenotypic evolution towards fosfomycin collateral sensitivity of Pseudomonas aeruginosa antibiotic-resistant mutants. Microb. Biotechnol. 2021. [Google Scholar] [CrossRef]
  55. Hancock, R.E.; Brinkman, F.S. Function of pseudomonas porins in uptake and efflux. Annu. Rev. Microbiol. 2002, 56, 17–38. [Google Scholar] [CrossRef] [Green Version]
  56. Wolter, D.J.; Lister, P.D. Mechanisms of beta-lactam resistance among Pseudomonas aeruginosa. Curr. Pharm. Des. 2013, 19, 209–222. [Google Scholar] [CrossRef]
  57. Aeschlimann, J.R. The role of multidrug efflux pumps in the antibiotic resistance of Pseudomonas aeruginosa and other gram-negative bacteria. Insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy 2003, 23, 916–924. [Google Scholar] [CrossRef]
  58. Torrens, G.; Hernandez, S.B.; Ayala, J.A.; Moya, B.; Juan, C.; Cava, F.; Oliver, A. Regulation of AmpC-Driven beta-Lactam Resistance in Pseudomonas aeruginosa: Different Pathways, Different Signaling. Msystems 2019, 4, e00524-19. [Google Scholar] [CrossRef] [Green Version]
  59. Ramirez, M.S.; Tolmasky, M.E. Aminoglycoside modifying enzymes. Drug Resist. Updat. 2010, 13, 151–171. [Google Scholar] [CrossRef] [Green Version]
  60. Fajardo, A.; Martinez-Martin, N.; Mercadillo, M.; Galan, J.C.; Ghysels, B.; Matthijs, S.; Cornelis, P.; Wiehlmann, L.; Tummler, B.; Baquero, F.; et al. The neglected intrinsic resistome of bacterial pathogens. PLoS ONE 2008, 3, e1619. [Google Scholar] [CrossRef] [Green Version]
  61. Linares, J.F.; Moreno, R.; Fajardo, A.; Martinez-Solano, L.; Escalante, R.; Rojo, F.; Martinez, J.L. The global regulator Crc modulates metabolism, susceptibility to antibiotics and virulence in Pseudomonas aeruginosa. Environ. Microbiol. 2010, 12, 3196–3212. [Google Scholar] [CrossRef]
  62. Merlo, C.A.; Boyle, M.P.; Diener-West, M.; Marshall, B.C.; Goss, C.H.; Lechtzin, N. Incidence and risk factors for multiple antibiotic-resistant Pseudomonas aeruginosa in cystic fibrosis. Chest 2007, 132, 562–568. [Google Scholar] [CrossRef]
  63. Llanes, C.; Hocquet, D.; Vogne, C.; Benali-Baitich, D.; Neuwirth, C.; Plesiat, P. Clinical strains of Pseudomonas aeruginosa overproducing MexAB-OprM and MexXY efflux pumps simultaneously. Antimicrob. Agents Chemother. 2004, 48, 1797–1802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yan, J.; Estanbouli, H.; Liao, C.; Kim, W.; Monk, J.M.; Rahman, R.; Kamboj, M.; Palsson, B.O.; Qiu, W.; Xavier, J.B. Systems-level analysis of NalD mutation, a recurrent driver of rapid drug resistance in acute Pseudomonas aeruginosa infection. PLoS Comput. Biol. 2019, 15, e1007562. [Google Scholar] [CrossRef] [PubMed]
  65. Jeannot, K.; Elsen, S.; Kohler, T.; Attree, I.; van Delden, C.; Plesiat, P. Resistance and virulence of Pseudomonas aeruginosa clinical strains overproducing the MexCD-OprJ efflux pump. Antimicrob. Agents Chemother. 2008, 52, 2455–2462. [Google Scholar] [CrossRef] [Green Version]
  66. Vettoretti, L.; Plesiat, P.; Muller, C.; El Garch, F.; Phan, G.; Attree, I.; Ducruix, A.; Llanes, C. Efflux unbalance in Pseudomonas aeruginosa isolates from cystic fibrosis patients. Antimicrob. Agents Chemother. 2009, 53, 1987–1997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Rodriguez-Martinez, J.M.; Poirel, L.; Nordmann, P. Extended-spectrum cephalosporinases in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2009, 53, 1766–1771. [Google Scholar] [CrossRef] [Green Version]
  68. Russ, D.; Glaser, F.; Shaer Tamar, E.; Yelin, I.; Baym, M.; Kelsic, E.D.; Zampaloni, C.; Haldimann, A.; Kishony, R. Escape mutations circumvent a tradeoff between resistance to a beta-lactam and resistance to a beta-lactamase inhibitor. Nat. Commun. 2020, 11, 2029. [Google Scholar] [CrossRef] [Green Version]
  69. Sanz-Garcia, F.; Hernando-Amado, S.; Martinez, J.L. Mutation-Driven Evolution of Pseudomonas aeruginosa in the Presence of either Ceftazidime or Ceftazidime-Avibactam. Antimicrob. Agents Chemother. 2018, 62, e01379-18. [Google Scholar] [CrossRef] [Green Version]
  70. Bruchmann, S.; Dotsch, A.; Nouri, B.; Chaberny, I.F.; Haussler, S. Quantitative contributions of target alteration and decreased drug accumulation to Pseudomonas aeruginosa fluoroquinolone resistance. Antimicrob. Agents Chemother. 2013, 57, 1361–1368. [Google Scholar] [CrossRef] [Green Version]
  71. Cabot, G.; Florit-Mendoza, L.; Sanchez-Diener, I.; Zamorano, L.; Oliver, A. Deciphering beta-lactamase-independent beta-lactam resistance evolution trajectories in Pseudomonas aeruginosa. J. Antimicrob. Chemother. 2018, 73, 3322–3331. [Google Scholar]
  72. Lopez-Causape, C.; Rubio, R.; Cabot, G.; Oliver, A. Evolution of the Pseudomonas aeruginosa Aminoglycoside Mutational Resistome In Vitro and in the Cystic Fibrosis Setting. Antimicrob. Agents Chemother. 2018, 62, e02583-17. [Google Scholar] [CrossRef] [Green Version]
  73. Breidenstein, E.B.; de la Fuente-Nunez, C.; Hancock, R.E. Pseudomonas aeruginosa: All roads lead to resistance. Trends Microbiol. 2011, 19, 419–426. [Google Scholar] [CrossRef]
  74. Botelho, J.; Grosso, F.; Peixe, L. Antibiotic resistance in Pseudomonas aeruginosa—Mechanisms, epidemiology and evolution. Drug Resist. Updat. 2019, 44, 100640. [Google Scholar] [CrossRef] [PubMed]
  75. Poole, K. Aminoglycoside resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2005, 49, 479–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Hong, D.J.; Bae, I.K.; Jang, I.H.; Jeong, S.H.; Kang, H.K.; Lee, K. Epidemiology and Characteristics of Metallo-β-Lactamase-Producing Pseudomonas aeruginosa. Infect. Chemother. 2015, 47, 81–97. [Google Scholar] [CrossRef] [PubMed]
  77. Ortiz de la Rosa, J.M.; Nordmann, P.; Poirel, L. Pathogenicity Genomic Island-Associated CrpP-Like Fluoroquinolone-Modifying Enzymes among Pseudomonas aeruginosa Clinical Isolates in Europe. Antimicrob. Agents Chemother. 2020, 64, e00489-20. [Google Scholar] [CrossRef]
  78. Laudy, A.; Róg, P.; Smolińska-Król, K.; Ćmiel, M.; Słoczyńska, A.; Patzer, J.; Dzierżanowska, D.; Wolinowska, R.; Starościak, B.; Tyski, S. Prevalence of ESBL-producing Pseudomonas aeruginosa isolates in Warsaw, Poland, detected by various phenotypic and genotypic methods. PLoS ONE 2017, 12, e0180121. [Google Scholar] [CrossRef] [Green Version]
  79. Cazares, A.; Moore, M.P.; Hall, J.P.J.; Wright, L.L.; Grimes, M.; Emond-Rheault, J.G.; Pongchaikul, P.; Santanirand, P.; Levesque, R.C.; Fothergill, J.L.; et al. A megaplasmid family driving dissemination of multidrug resistance in Pseudomonas. Nat. Commun. 2020, 11, 1370. [Google Scholar] [CrossRef] [Green Version]
  80. Liapis, E.; Bour, M.; Triponney, P.; Jove, T.; Zahar, J.R.; Valot, B.; Jeannot, K.; Plesiat, P. Identification of Diverse Integron and Plasmid Structures Carrying a Novel Carbapenemase Among Pseudomonas Species. Front. Microbiol. 2019, 10, 404. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, M.; Ma, J.; Jia, W.; Li, W. Antimicrobial Resistance and Molecular Characterization of Gene Cassettes from Class 1 Integrons in Pseudomonas aeruginosa Strains. Microb. Drug Resist. 2020, 26, 670–676. [Google Scholar] [CrossRef]
  82. Poirel, L.; Lambert, T.; Turkoglu, S.; Ronco, E.; Gaillard, J.; Nordmann, P. Characterization of Class 1 integrons from Pseudomonas aeruginosa that contain the bla(VIM-2) carbapenem-hydrolyzing beta-lactamase gene and of two novel aminoglycoside resistance gene cassettes. Antimicrob. Agents Chemother. 2001, 45, 546–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Van der Zee, A.; Kraak, W.B.; Burggraaf, A.; Goessens, W.H.F.; Pirovano, W.; Ossewaarde, J.M.; Tommassen, J. Spread of Carbapenem Resistance by Transposition and Conjugation Among Pseudomonas aeruginosa. Front. Microbiol. 2018, 9, 2057. [Google Scholar] [CrossRef]
  84. Brown-Jaque, M.; Rodriguez Oyarzun, L.; Cornejo-Sánchez, T.; Martín-Gómez, M.T.; Gartner, S.; de Gracia, J.; Rovira, S.; Alvarez, A.; Jofre, J.; González-López, J.J.; et al. Detection of Bacteriophage Particles Containing Antibiotic Resistance Genes in the Sputum of Cystic Fibrosis Patients. Front. Microbiol. 2018, 9, 856. [Google Scholar] [CrossRef] [PubMed]
  85. Galetti, R.; Andrade, L.N.; Varani, A.M.; Darini, A.L.C. A Phage-Like Plasmid Carrying blaKPC-2 Gene in Carbapenem-Resistant Pseudomonas aeruginosa. Front. Microbiol. 2019, 10, 572. [Google Scholar] [CrossRef] [PubMed]
  86. Sanz-Garcia, F.; Alvarez-Ortega, C.; Olivares-Pacheco, J.; Blanco, P.; Martinez, J.L.; Hernando-Amado, S. Analysis of the Pseudomonas aeruginosa Aminoglycoside Differential Resistomes Allows Defining Genes Simultaneously Involved in Intrinsic Antibiotic Resistance and Virulence. Antimicrob. Agents Chemother. 2019, 63, e00185-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Sanz-García, F.; Hernando-Amado, S.; Martínez, J.L. Evolution under low antibiotic concentrations: A risk for the selection of Pseudomonas aeruginosa multidrug resistant mutants in nature. bioRxiv 2021. [Google Scholar] [CrossRef]
  88. Danner, M.C.; Robertson, A.; Behrends, V.; Reiss, J. Antibiotic pollution in surface fresh waters: Occurrence and effects. Sci. Total Environ. 2019, 664, 793–804. [Google Scholar] [CrossRef]
  89. Martinez, J.L. Antibiotics and antibiotic resistance genes in natural environments. Science 2008, 321, 365–367. [Google Scholar] [CrossRef] [PubMed]
  90. Costerton, J.W.; Stewart, P.S.; Greenberg, E.P. Bacterial biofilms: A common cause of persistent infections. Science 1999, 284, 1318–1322. [Google Scholar] [CrossRef] [Green Version]
  91. Mc, D.W. Microbial persistence. Yale J. Biol. Med. 1958, 30, 257–291. [Google Scholar]
  92. Nguyen, D.; Joshi-Datar, A.; Lepine, F.; Bauerle, E.; Olakanmi, O.; Beer, K.; McKay, G.; Siehnel, R.; Schafhauser, J.; Wang, Y.; et al. Active starvation responses mediate antibiotic tolerance in biofilms and nutrient-limited bacteria. Science 2011, 334, 982–986. [Google Scholar] [CrossRef] [Green Version]
  93. Moker, N.; Dean, C.R.; Tao, J. Pseudomonas aeruginosa increases formation of multidrug-tolerant persister cells in response to quorum-sensing signaling molecules. J. Bacteriol. 2010, 192, 1946–1955. [Google Scholar] [CrossRef] [Green Version]
  94. Lewis, K. Persister cells and the riddle of biofilm survival. Biochemistry 2005, 70, 267–274. [Google Scholar] [CrossRef]
  95. Sadovskaya, I.; Vinogradov, E.; Li, J.; Hachani, A.; Kowalska, K.; Filloux, A. High-level antibiotic resistance in Pseudomonas aeruginosa biofilm: The ndvB gene is involved in the production of highly glycerol-phosphorylated beta-(1->3)-glucans, which bind aminoglycosides. Glycobiology 2010, 20, 895–904. [Google Scholar] [CrossRef] [Green Version]
  96. Pamp, S.J.; Gjermansen, M.; Johansen, H.K.; Tolker-Nielsen, T. Tolerance to the antimicrobial peptide colistin in Pseudomonas aeruginosa biofilms is linked to metabolically active cells, and depends on the pmr and mexAB-oprM genes. Mol. Microbiol. 2008, 68, 223–240. [Google Scholar] [CrossRef] [PubMed]
  97. Tata, M.; Wolfinger, M.T.; Amman, F.; Roschanski, N.; Dotsch, A.; Sonnleitner, E.; Haussler, S.; Blasi, U. RNASeq Based Transcriptional Profiling of Pseudomonas aeruginosa PA14 after Short- and Long-Term Anoxic Cultivation in Synthetic Cystic Fibrosis Sputum Medium. PLoS ONE 2016, 11, e0147811. [Google Scholar] [CrossRef]
  98. Kubicek-Sutherland, J.Z.; Heithoff, D.M.; Ersoy, S.C.; Shimp, W.R.; House, J.K.; Marth, J.D.; Smith, J.W.; Mahan, M.J. Host-dependent Induction of Transient Antibiotic Resistance: A Prelude to Treatment Failure. EBioMedicine 2015, 2, 1169–1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Hanson, N.D.; Sanders, C.C. Regulation of inducible AmpC beta-lactamase expression among Enterobacteriaceae. Curr. Pharm. Des. 1999, 5, 881–894. [Google Scholar]
  100. Groisman, E.A.; Kayser, J.; Soncini, F.C. Regulation of polymyxin resistance and adaptation to low-Mg2+ environments. J. Bacteriol. 1997, 179, 7040–7045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Fernandez, L.; Gooderham, W.J.; Bains, M.; McPhee, J.B.; Wiegand, I.; Hancock, R.E. Adaptive resistance to the “last hope” antibiotics polymyxin B and colistin in Pseudomonas aeruginosa is mediated by the novel two-component regulatory system ParR-ParS. Antimicrob. Agents Chemother. 2010, 54, 3372–3382. [Google Scholar] [CrossRef] [Green Version]
  102. Corona, F.; Martinez, J.L. Phenotypic Resistance to Antibiotics. Antibiotics 2013, 2, 237–255. [Google Scholar] [CrossRef] [Green Version]
  103. Laborda, P.; Alcalde-Rico, M.; Blanco, P.; Martínez, J.L.; Hernando-Amado, S. Novel Inducers of the Expression of Multidrug Efflux Pumps That Trigger Pseudomonas aeruginosa Transient Antibiotic Resistance. Antimicrob. Agents Chemother. 2019, 63, e01095-19. [Google Scholar] [CrossRef]
  104. Strempel, N.; Neidig, A.; Nusser, M.; Geffers, R.; Vieillard, J.; Lesouhaitier, O.; Brenner-Weiss, G.; Overhage, J. Human host defense peptide LL-37 stimulates virulence factor production and adaptive resistance in Pseudomonas aeruginosa. PLoS ONE 2013, 8, e82240. [Google Scholar] [CrossRef]
  105. Chen, H.; Hu, J.; Chen, P.R.; Lan, L.; Li, Z.; Hicks, L.M.; Dinner, A.R.; He, C. The Pseudomonas aeruginosa multidrug efflux regulator MexR uses an oxidation-sensing mechanism. Proc. Natl. Acad. Sci. USA 2008, 105, 13586–13591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Ghosh, S.; Cremers, C.M.; Jakob, U.; Love, N.G. Chlorinated phenols control the expression of the multidrug resistance efflux pump MexAB-OprM in Pseudomonas aeruginosa by interacting with NalC. Mol. Microbiol. 2011, 79, 1547–1556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Fetar, H.; Gilmour, C.; Klinoski, R.; Daigle, D.M.; Dean, C.R.; Poole, K. mexEF-oprN multidrug efflux operon of Pseudomonas aeruginosa: Regulation by the MexT activator in response to nitrosative stress and chloramphenicol. Antimicrob. Agents Chemother. 2011, 55, 508–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Frisk, A.; Schurr, J.R.; Wang, G.; Bertucci, D.C.; Marrero, L.; Hwang, S.H.; Hassett, D.J.; Schurr, M.J. Transcriptome analysis of Pseudomonas aeruginosa after interaction with human airway epithelial cells. Infect. Immun. 2004, 72, 5433–5438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Morita, Y.; Tomida, J.; Kawamura, Y. MexXY multidrug efflux system of Pseudomonas aeruginosa. Front. Microbiol. 2012, 3, 408. [Google Scholar] [CrossRef] [Green Version]
  110. Levin-Reisman, I.; Brauner, A.; Ronin, I.; Balaban, N.Q. Epistasis between antibiotic tolerance, persistence, and resistance mutations. Proc. Natl. Acad. Sci. USA 2019, 116, 14734–14739. [Google Scholar] [CrossRef] [Green Version]
  111. Santi, I.; Manfredi, P.; Maffei, E.; Egli, A.; Jenal, U. Evolution of Antibiotic Tolerance Shapes Resistance Development in Chronic Pseudomonas aeruginosa Infections. Mbio 2021, 12, e03482-20. [Google Scholar] [CrossRef] [PubMed]
  112. Carpenter, R.J.; Hartzell, J.D.; Forsberg, J.A.; Babel, B.S.; Ganesan, A. Pseudomonas putida war wound infection in a US Marine: A case report and review of the literature. J. Infect. 2008, 56, 234–240. [Google Scholar] [CrossRef] [PubMed]
  113. Chamon, R.C.; Abel da Rocha, J.; Araujo Martins, I.; Lopes Pires, L.; Macêdo de Almeida, B.; Souza Leite, N.; Rezende Vieira de Mendonça Souza, C.; Zahner, V.; Leite Ribeiro, R.; Pavoni Gomes Chagas, T.; et al. KPC-2 producing Pseudomonas putida as an unexpected pathogen of catheter-associated bloodstream infection. J. Infect. Dev. Ctries. 2020, 14, 411–414. [Google Scholar] [CrossRef] [PubMed]
  114. Tan, G.; Xi, Y.; Yuan, P.; Sun, Z.; Yang, D. Risk factors and antimicrobial resistance profiles of Pseudomonas putida infection in Central China, 2010–2017. Medicine 2019, 98, e17812. [Google Scholar] [CrossRef]
  115. Usta Atmaca, H.; Akbas, F. A Extensively drug-resistant Pseudomonas putida bacteremia that was resolved spontaneously. J. Infect. Dev. Ctries. 2019, 13, 577–580. [Google Scholar] [CrossRef]
  116. Fernández, M.; Porcel, M.; de la Torre, J.; Molina-Henares, M.A.; Daddaoua, A.; Llamas, M.A.; Roca, A.; Carriel, V.; Garzón, I.; Ramos, J.L.; et al. Analysis of the pathogenic potential of nosocomial Pseudomonas putida strains. Front. Microbiol. 2015, 6, 871. [Google Scholar] [CrossRef] [PubMed]
  117. Molina, L.; Udaondo, Z.; Duque, E.; Fernandez, M.; Bernal, P.; Roca, A.; de la Torre, J.; Ramos, J.L. Specific Gene Loci of Clinical Pseudomonas putida Isolates. PLoS ONE 2016, 11, e0147478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Towner, K.J. Acinetobacter: An old friend, but a new enemy. J. Hosp. Infect. 2009, 73, 355–363. [Google Scholar] [CrossRef]
  119. Howard, A.; O’Donoghue, M.; Feeney, A.; Sleator, R.D. Acinetobacterbaumannii: An emerging opportunistic pathogen. Virulence 2012, 3, 243–250. [Google Scholar] [CrossRef]
  120. Eveillard, M.; Kempf, M.; Belmonte, O.; Pailhoriès, H.; Joly-Guillou, M.-L. Reservoirs of Acinetobacter baumannii outside the hospital and potential involvement in emerging human community-acquired infections. Int. J. Infect. Dis. 2013, 17, e802–e805. [Google Scholar] [CrossRef] [Green Version]
  121. Touchon, M.; Cury, J.; Yoon, E.J.; Krizova, L.; Cerqueira, G.C.; Murphy, C.; Feldgarden, M.; Wortman, J.; Clermont, D.; Lambert, T.; et al. The genomic diversification of the whole Acinetobacter genus: Origins, mechanisms, and consequences. Genome Biol. Evol. 2014, 6, 2866–2882. [Google Scholar] [CrossRef] [Green Version]
  122. Chan, A.P.; Sutton, G.; DePew, J.; Krishnakumar, R.; Choi, Y.; Huang, X.Z.; Beck, E.; Harkins, D.M.; Kim, M.; Lesho, E.P.; et al. A novel method of consensus pan-chromosome assembly and large-scale comparative analysis reveal the highly flexible pan-genome of Acinetobacter baumannii. Genome Biol. 2015, 16, 143. [Google Scholar] [CrossRef] [Green Version]
  123. Figueiredo, S.; Bonnin, R.A.; Poirel, L.; Duranteau, J.; Nordmann, P. Identification of the naturally occurring genes encoding carbapenem-hydrolysing oxacillinases from Acinetobacter haemolyticus, Acinetobacter johnsonii, and Acinetobacter calcoaceticus. Clin. Microbiol. Infect. 2012, 18, 907–913. [Google Scholar] [CrossRef] [Green Version]
  124. Antunes, L.C.S.; Imperi, F.; Carattoli, A.; Visca, P. Deciphering the Multifactorial Nature of Acinetobacter baumannii Pathogenicity. PLoS ONE 2011, 6, e22674. [Google Scholar] [CrossRef] [Green Version]
  125. Karah, N.; Sundsfjord, A.; Towner, K.; Samuelsen, Ø. Insights into the global molecular epidemiology of carbapenem non-susceptible clones of Acinetobacter baumannii. Drug Resist. Updates 2012, 15, 237–247. [Google Scholar] [CrossRef]
  126. Diancourt, L.; Passet, V.; Nemec, A.; Dijkshoorn, L.; Brisse, S. The Population Structure of Acinetobacter baumannii: Expanding Multiresistant Clones from an Ancestral Susceptible Genetic Pool. PLoS ONE 2010, 5, e10034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Mangas, E.L.; Rubio, A.; Álvarez-Marín, R.; Labrador-Herrera, G.; Pachón, J.; Pachón-Ibáñez, M.E.; Divina, F.; Pérez-Pulido, A.J. Pangenome of Acinetobacter baumannii uncovers two groups of genomes, one of them with genes involved in CRISPR/Cas defence systems associated with the absence of plasmids and exclusive genes for biofilm formation. Microb. Genom. 2019, 5, e000309. [Google Scholar] [CrossRef]
  128. Kanafani, Z.A.; Zahreddine, N.; Tayyar, R.; Sfeir, J.; Araj, G.F.; Matar, G.M.; Kanj, S.S. Multi-drug resistant Acinetobacter species: A seven-year experience from a tertiary care center in Lebanon. Antimicrob. Resist. Infect. Control. 2018, 7, 9. [Google Scholar] [CrossRef] [Green Version]
  129. Zordan, S. Multidrug-Resistant Acinetobacter baumannii in Veterinary Clinics, Germany. Emerg. Infect. Dis. 2011, 17, 1751–1754. [Google Scholar] [CrossRef] [PubMed]
  130. Hernández-González, I.L.; Castillo-Ramírez, S. Antibiotic-resistant Acinetobacter baumannii is a One Health problem. Lancet Microbe 2020, 1, e279. [Google Scholar] [CrossRef]
  131. Bou, G.; Martínez-Beltrán, J. Cloning, nucleotide sequencing, and analysis of the gene encoding an AmpC beta-lactamase in Acinetobacter baumannii. Antimicrob. Agents Chemother. 2000, 44, 428–432. [Google Scholar] [CrossRef] [Green Version]
  132. Magnet, S.; Courvalin, P.; Lambert, T. Resistance-nodulation-cell division-type efflux pump involved in aminoglycoside resistance in Acinetobacter baumannii strain BM4454. Antimicrob. Agents Chemother. 2001, 45, 3375–3380. [Google Scholar] [CrossRef] [Green Version]
  133. Damier-Piolle, L.; Magnet, S.; Bremont, S.; Lambert, T.; Courvalin, P. AdeIJK, a Resistance-Nodulation-Cell Division Pump Effluxing Multiple Antibiotics in Acinetobacter baumannii. Antimicrob. Agents Chemother. 2008, 52, 557–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Coyne, S.; Courvalin, P.; Périchon, B. Efflux-mediated antibiotic resistance in Acinetobacter spp. Antimicrob. Agents Chemother. 2011, 55, 947–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kim, Y.; Bae, I.; Lee, H.; Yong, D.; Lee, K. In vivo emergence of colistin resistance in Acinetobacter baumannii clinical isolates of sequence type 357 during colistin treatment. Diagn. Microbiol. Infect. Dis. 2014, 79, 362–366. [Google Scholar] [CrossRef]
  136. Lucaßen, K.; Müller, C.; Wille, J.; Xanthopoulou, K.; Hackel, M.; Seifert, H.; Higgins, P.G. Prevalence of RND efflux pump regulator variants associated with tigecycline resistance in carbapenem-resistant Acinetobacter baumannii from a worldwide survey. J. Antimicrob. Chemother. 2021, 76, 1724–1730. [Google Scholar] [CrossRef]
  137. Uppalapati, S.R.; Sett, A.; Pathania, R. The Outer Membrane Proteins OmpA, CarO, and OprD of Acinetobacter baumannii Confer a Two-Pronged Defense in Facilitating Its Success as a Potent Human Pathogen. Front. Microbiol. 2020, 11, 589234. [Google Scholar] [CrossRef]
  138. Martinez, J.L.; Baquero, F. Interactions among strategies associated with bacterial infection: Pathogenicity, epidemicity, and antibiotic resistance. Clin. Microbiol. Rev. 2002, 15, 647–679. [Google Scholar] [CrossRef] [Green Version]
  139. Ramirez, M.S.; Bonomo, R.A.; Tolmasky, M.E. Carbapenemases: Transforming Acinetobacter baumannii into a Yet More Dangerous Menace. Biomolecules 2020, 10, 720. [Google Scholar] [CrossRef] [PubMed]
  140. Hujer, A.M.; Hujer, K.M.; Leonard, D.A.; Powers, R.A.; Wallar, B.J.; Mack, A.R.; Taracila, M.A.; Rather, P.N.; Higgins, P.G.; Prati, F.; et al. A comprehensive and contemporary “snapshot” of β-lactamases in carbapenem resistant Acinetobacter baumannii. Diagn. Microbiol. Infect. Dis. 2021, 99, 115242. [Google Scholar] [CrossRef]
  141. Słoczyńska, A.; Wand, M.E.; Tyski, S.; Laudy, A.E. Analysis of bla(CHDL) Genes and Insertion Sequences Related to Carbapenem Resistance in Acinetobacter baumannii Clinical Strains Isolated in Warsaw, Poland. Int. J. Mol. Sci. 2021, 22, 2486. [Google Scholar] [CrossRef]
  142. Chamoun, S.; Welander, J.; Martis-Thiele, M.M.; Ntzouni, M.; Claesson, C.; Vikström, E.; Turkina, M.V. Colistin Dependence in Extensively Drug-Resistant Acinetobacter baumannii Strain Is Associated with ISAjo2 and ISAba13 Insertions and Multiple Cellular Responses. Int. J. Mol. Sci. 2021, 22, 576. [Google Scholar] [CrossRef]
  143. Vijayakumar, S.; Anandan, S.; Prabaa, D.; Kanthan, K.; Vijayabaskar, S.; Kapil, A.; Ray, P.; Sistla, S.; Bhattacharya, S.; Wattal, C.; et al. Insertion sequences and sequence types profile of clinical isolates of carbapenem-resistant A. baumannii collected across India over four year period. J. Infect. Public Health 2020, 13, 1022–1028. [Google Scholar] [CrossRef] [PubMed]
  144. Cerezales, M.; Xanthopoulou, K.; Wille, J.; Krut, O.; Seifert, H.; Gallego, L.; Higgins, P.G. Mobile Genetic Elements Harboring Antibiotic Resistance Determinants in Acinetobacter baumannii Isolates From Bolivia. Front. Microbiol. 2020, 11, 919. [Google Scholar] [CrossRef]
  145. Adams, M.D.; Chan, E.R.; Molyneaux, N.D.; Bonomo, R.A. Genomewide Analysis of Divergence of Antibiotic Resistance Determinants in Closely Related Isolates of Acinetobacter baumannii. Antimicrob. Agents Chemother. 2010, 54, 3569. [Google Scholar] [CrossRef] [Green Version]
  146. Fournier, P.E.; Vallenet, D.; Barbe, V.; Audic, S.; Ogata, H.; Poirel, L.; Richet, H.; Robert, C.; Mangenot, S.; Abergel, C.; et al. Comparative Genomics of Multidrug Resistance in Acinetobacter baumannii. PLoS Genet. 2006, 2, e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Pagano, M.; Martins, A.F.; Machado, A.B.M.P.; Barin, J.; Barth, A.L. Carbapenem-susceptible Acinetobacter baumannii carrying the ISAba1 upstream blaOXA-51-like gene in Porto Alegre, southern Brazil. Epidemiol. Infect. 2013, 141, 330–333. [Google Scholar] [CrossRef] [PubMed]
  148. Fonseca, É.L.; Freitas, F.d.S.; Scheidegger, É.M.D.; Jacinto, T.; Vicente, A.C.P. Class 2 integrons in multidrug-resistant Acinetobacter baumannii circulating in different Brazilian geographic regions. Int. J. Antimicrob. Agents 2011, 38, 95–96. [Google Scholar] [CrossRef]
  149. Potron, A.; Poirel, L.; Nordmann, P. Emerging broad-spectrum resistance in Pseudomonas aeruginosa and Acinetobacter baumannii: Mechanisms and epidemiology. Int. J. Antimicrob. Agents 2015, 45, 568–585. [Google Scholar] [CrossRef] [Green Version]
  150. Bi, D.; Zheng, J.; Xie, R.; Zhu, Y.; Wei, R.; Ou, H.Y.; Wei, Q.; Qin, H. Comparative Analysis of AbaR-Type Genomic Islands Reveals Distinct Patterns of Genetic Features in Elements with Different Backbones. Msphere 2020, 5, e00349-20. [Google Scholar] [CrossRef]
  151. Bi, D.; Xie, R.; Zheng, J.; Yang, H.; Zhu, X.; Ou, H.-Y.; Wei, Q. Large-Scale Identification of AbaR-Type Genomic Islands in Acinetobacter baumannii Reveals Diverse Insertion Sites and Clonal Lineage-Specific Antimicrobial Resistance Gene Profiles. Antimicrob. Agents Chemother. 2019, 63, e02526-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Cameranesi, M.M.; Paganini, J.; Limansky, A.S.; Moran-Barrio, J.; Salcedo, S.P.; Viale, A.M.; Repizo, G.D. Acquisition of plasmids conferring carbapenem and aminoglycoside resistance and loss of surface-exposed macromolecule structures as strategies for the adaptation of Acinetobacter baumannii CC104(O)/CC15(P) strains to the clinical setting. Microb. Genom. 2020, 6. [Google Scholar] [CrossRef]
  153. Wachino, J.I.; Jin, W.; Kimura, K.; Arakawa, Y. Intercellular Transfer of Chromosomal Antimicrobial Resistance Genes between Acinetobacter baumannii Strains Mediated by Prophages. Antimicrob. Agents Chemother. 2019, 63, e00334-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Krahn, T.; Wibberg, D.; Maus, I.; Winkler, A.; Bontron, S.; Sczyrba, A.; Nordmann, P.; Pühler, A.; Poirel, L.; Schlüter, A. Intraspecies Transfer of the Chromosomal Acinetobacter baumannii blaNDM-1 Carbapenemase Gene. Antimicrob. Agents Chemother. 2016, 60, 3032–3040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Vesel, N.; Blokesch, M. Pilus Production in Acinetobacter baumannii Is Growth Phase Dependent and Essential for Natural Transformation. J. Bacteriol. 2021, 203, e00034-21. [Google Scholar] [CrossRef]
  156. Ramirez, M.S.; Don, M.; Merkier, A.K.; Bistué, A.J.; Zorreguieta, A.; Centrón, D.; Tolmasky, M.E. Naturally competent Acinetobacter baumannii clinical isolate as a convenient model for genetic studies. J. Clin. Microbiol. 2010, 48, 1488–1490. [Google Scholar] [CrossRef] [Green Version]
  157. Evans, B.A.; Kumar, A.; Castillo-Ramírez, S. Editorial: Genomic Basis of Antibiotic Resistance and Virulence in Acinetobacter. Front. Microbiol. 2021, 12, 670975. [Google Scholar] [CrossRef]
  158. Weber, B.S.; Harding, C.M.; Feldman, M.F. Pathogenic Acinetobacter: From the Cell Surface to Infinity and Beyond. J. Bacteriol. 2016, 198, 880–887. [Google Scholar] [CrossRef] [Green Version]
  159. Zhang, L.; Fu, Y.; Han, X.; Xu, Q.; Weng, S.; Yan, B.; Liu, L.; Hua, X.; Chen, Y.; Yu, Y. Phenotypic Variation and Carbapenem Resistance Potential in OXA-499-Producing Acinetobacter pittii. Front. Microbiol. 2020, 11, 1134. [Google Scholar] [CrossRef]
  160. Knight, D.B.; Rudin, S.D.; Bonomo, R.A.; Rather, P.N. Acinetobacter nosocomialis: Defining the Role of Efflux Pumps in Resistance to Antimicrobial Therapy, Surface Motility, and Biofilm Formation. Front. Microbiol. 2018, 9, 1902. [Google Scholar] [CrossRef]
  161. Kim, D.; Baik, K.S.; Kim, M.S.; Park, S.C.; Kim, S.S.; Rhee, M.S.; Kwak, Y.S.; Seong, C.N. Acinetobactersoli sp. nov., isolated from forest soil. J. Microbiol. 2008, 46, 396–401. [Google Scholar] [CrossRef]
  162. Kumsa, B.; Socolovschi, C.; Parola, P.; Rolain, J.-M.; Raoult, D. Molecular Detection of Acinetobacter Species in Lice and Keds of Domestic Animals in Oromia Regional State, Ethiopia. PLoS ONE 2012, 7, e52377. [Google Scholar] [CrossRef] [Green Version]
  163. Pellegrino, F.L.P.C.; Vieira, V.V.; Baio, P.V.P.; dos Santos, R.M.R.; dos Santos, A.L.A.; Santos, N.G.d.B.; Meohas, M.M.G.L.; Santos, R.T.; de Souza, T.C.; da Silva Dias, R.C.; et al. Acinetobactersoli as a Cause of Bloodstream Infection in a Neonatal Intensive Care Unit: Table 1. J. Clin. Microbiol. 2011, 49, 2283–2285. [Google Scholar] [CrossRef] [Green Version]
  164. Cao, J.-G.; Zhang, H.; Xu, Y.-P.; Yan, Z.-Q.; Liu, Y.-X. First report of carbapenem-resistant Acinetobacter soli isolates coharboring blaNDM-1 and blaOXA-58 genes from China. Diagn. Microbiol. Infect. Dis. 2015, 83, 407–408. [Google Scholar] [CrossRef] [PubMed]
  165. Toleman, M.A.; Spencer, J.; Jones, L.; Walsh, T.R. blaNDM-1 is a chimera likely constructed in Acinetobacter baumannii. Antimicrob. Agents Chemother. 2012, 56, 2773–2776. [Google Scholar] [CrossRef] [Green Version]
  166. Sung, J.Y.; Koo, S.H.; Kim, S.; Kwon, K.C. Epidemiological Characterizations of Class 1 Integrons from Multidrug-Resistant Acinetobacter Isolates in Daejeon, Korea. Ann. Lab. Med. 2014, 34, 293–299. [Google Scholar] [CrossRef]
  167. Huang, T.-W.; Lauderdale, T.-L.; Liao, T.-L.; Hsu, M.-C.; Chang, F.-Y.; Chang, S.-C.; Khong, W.X.; Ng, O.T.; Chen, Y.-T.; Kuo, S.-C.; et al. Effective transfer of a 47 kb NDM-1-positive plasmid among Acinetobacter species. J. Antimicrob. Chemother. 2015, 70, 2734–2738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Silva, L.; Mourão, J.; Grosso, F.; Peixe, L. Uncommon carbapenemase-encoding plasmids in the clinically emergent Acinetobacter pittii. J. Antimicrob. Chemother. 2018, 73, 52–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Salto, I.P.; Torres Tejerizo, G.; Wibberg, D.; Pühler, A.; Schlüter, A.; Pistorio, M. Comparative genomic analysis of Acinetobacter spp. plasmids originating from clinical settings and environmental habitats. Sci. Rep. 2018, 8, 7783. [Google Scholar] [CrossRef] [Green Version]
  170. Salgado-Camargo, A.D.; Castro-Jaimes, S.; Gutierrez-Rios, R.-M.; Lozano, L.F.; Altamirano-Pacheco, L.; Silva-Sanchez, J.; Pérez-Oseguera, Á.; Volkow, P.; Castillo-Ramírez, S.; Cevallos, M.A. Structure and Evolution of Acinetobacter baumannii Plasmids. Front. Microbiol. 2020, 11, 1283. [Google Scholar] [CrossRef] [PubMed]
  171. Nemec, A.; Musílek, M.; Šedo, O.; De Baere, T.; Maixnerová, M.; van der Reijden, T.J.K.; Zdráhal, Z.; Vaneechoutte, M.; Dijkshoorn, L. Acinetobacter bereziniae sp. nov. and Acinetobacter guillouiae sp. nov., to accommodate Acinetobacter genomic species 10 and 11, respectively. Int. J. Syst. Evol. Microbiol. 2010, 60, 896–903. [Google Scholar] [CrossRef] [Green Version]
  172. Zander, E.; Fernández-González, A.; Schleicher, X.; Dammhayn, C.; Kamolvit, W.; Seifert, H.; Higgins, P.G. Worldwide dissemination of acquired carbapenem-hydrolysing class D β-lactamases in Acinetobacter spp. other than Acinetobacter baumannii. Int. J. Antimicrob. Agents 2014, 43, 375–377. [Google Scholar] [CrossRef] [PubMed]
  173. Brovedan, M.; Marchiaro, P.M.; Morán-Barrio, J.; Revale, S.; Cameranesi, M.; Brambilla, L.; Viale, A.M.; Limansky, A.S. Draft Genome Sequence of Acinetobacter bereziniae HPC229, a Carbapenem-Resistant Clinical Strain from Argentina Harboring blaNDM-1. Genome Announc. 2016, 4, e00117-16. [Google Scholar] [CrossRef] [Green Version]
  174. Regeen, H.; Al-Sharafa-Kittaneh, D.; Kattan, R.; Al-Dawodi, R.; Marzouqa, H.; Hindiyeh, M.Y. First Report of blaNDM and blaOXA-58 Coexistence in Acinetobacter junii: TABLE 1. J. Clin. Microbiol. 2014, 52, 3492–3493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Brooke, J.S. Stenotrophomonas maltophilia: An emerging global opportunistic pathogen. Clin. Microbiol. Rev. 2012, 25, 2–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Juhnke, M.E.; des Jardin, E. Selective medium for isolation of Xanthomonas maltophilia from soil and rhizosphere environments. Appl. Environ. Microbiol. 1989, 55, 747–750. [Google Scholar] [CrossRef] [Green Version]
  177. Dungan, R.S.; Yates, S.R.; Frankenberger, W.T., Jr. Transformations of selenate and selenite by Stenotrophomonas maltophilia isolated from a seleniferous agricultural drainage pond sediment. Environ. Microbiol. 2003, 5, 287–295. [Google Scholar] [CrossRef]
  178. Ochoa-Sanchez, L.E.; Vinuesa, P. Evolutionary Genetic Analysis Uncovers Multiple Species with Distinct Habitat Preferences and Antibiotic Resistance Phenotypes in the Stenotrophomonas maltophilia Complex. Front. Microbiol. 2017, 8, 1548. [Google Scholar] [CrossRef] [Green Version]
  179. Matyar, F.; Kaya, A.; Dincer, S. Antibacterial agents and heavy metal resistance in Gram-negative bacteria isolated from seawater, shrimp and sediment in Iskenderun Bay, Turkey. Sci. Total Environ. 2008, 407, 279–285. [Google Scholar] [CrossRef] [PubMed]
  180. Berg, G.; Martinez, J.L. Friends or foes: Can we make a distinction between beneficial and harmful strains of the Stenotrophomonas maltophilia complex? Front. Microbiol. 2015, 6, 241. [Google Scholar] [CrossRef]
  181. Gröschel, M.I.; Meehan, C.J.; Barilar, I.; Diricks, M.; Gonzaga, A.; Steglich, M.; Conchillo-Solé, O.; Scherer, I.C.; Mamat, U.; Luz, C.F.; et al. The phylogenetic landscape and nosocomial spread of the multidrug-resistant opportunist Stenotrophomonas maltophilia. Nat. Commun. 2020, 11, 2044. [Google Scholar] [CrossRef]
  182. Morales, G.; Wiehlmann, L.; Gudowius, P.; van Delden, C.; Tummler, B.; Martinez, J.L.; Rojo, F. Structure of Pseudomonas aeruginosa populations analyzed by single nucleotide polymorphism and pulsed-field gel electrophoresis genotyping. J. Bacteriol. 2004, 186, 4228–4237. [Google Scholar] [CrossRef] [Green Version]
  183. Alonso, A.; Rojo, F.; Martinez, J.L. Environmental and clinical isolates of Pseudomonas aeruginosa show pathogenic and biodegradative properties irrespective of their origin. Environ. Microbiol. 1999, 1, 421–430. [Google Scholar] [CrossRef] [PubMed]
  184. Lira, F.; Berg, G.; Martinez, J.L. Double-Face Meets the Bacterial World: The Opportunistic Pathogen Stenotrophomonas maltophilia. Front. Microbiol. 2017, 8, 2190. [Google Scholar] [CrossRef] [PubMed]
  185. Brooke, J.S.; Di Bonaventura, G.; Berg, G.; Martinez, J.L. Editorial: A Multidisciplinary Look at Stenotrophomonas maltophilia: An Emerging Multi-Drug-Resistant Global Opportunistic Pathogen. Front. Microbiol. 2017, 8, 1511. [Google Scholar] [CrossRef] [PubMed]
  186. Svensson-Stadler, L.A.; Mihaylova, S.A.; Moore, E.R.B. Stenotrophomonas interspecies differentiation and identification by gyrB sequence analysis. FEMS Microbiol. Lett. 2012, 327, 15–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Adamek, M.; Overhage, J.; Bathe, S.; Winter, J.; Fischer, R.; Schwartz, T. Genotyping of environmental and clinical Stenotrophomonas maltophilia isolates and their pathogenic potential. PLoS ONE 2011, 6, e27615. [Google Scholar] [CrossRef] [Green Version]
  188. Vasileuskaya-Schulz, Z.; Kaiser, S.; Maier, T.; Kostrzewa, M.; Jonas, D. Delineation of Stenotrophomonas spp. by multi-locus sequence analysis and MALDI-TOF mass spectrometry. Syst. Appl. Microbiol. 2011, 34, 35–39. [Google Scholar] [CrossRef]
  189. Valdezate, S.; Vindel, A.; Martin-Davila, P.; Del Saz, B.S.; Baquero, F.; Canton, R. High genetic diversity among Stenotrophomonas maltophilia strains despite their originating at a single hospital. J. Clin. Microbiol. 2004, 42, 693–699. [Google Scholar] [CrossRef] [Green Version]
  190. Deredjian, A.; Alliot, N.; Blanchard, L.; Brothier, E.; Anane, M.; Cambier, P.; Jolivet, C.; Khelil, M.N.; Nazaret, S.; Saby, N.; et al. Occurrence of Stenotrophomonas maltophilia in agricultural soils and antibiotic resistance properties. Res. Microbiol. 2016, 167, 313–324. [Google Scholar] [CrossRef]
  191. Vinuesa, P.; Ochoa-Sanchez, L.E.; Contreras-Moreira, B. GET_PHYLOMARKERS, a Software Package to Select Optimal Orthologous Clusters for Phylogenomics and Inferring Pan-Genome Phylogenies, Used for a Critical Geno-Taxonomic Revision of the Genus Stenotrophomonas. Front. Microbiol. 2018, 9, 771. [Google Scholar] [CrossRef] [Green Version]
  192. Patil, P.P.; Midha, S.; Kumar, S.; Patil, P.B. Genome Sequence of Type Strains of Genus Stenotrophomonas. Front. Microbiol. 2016, 7, 309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Senol, E. Stenotrophomonas maltophilia: The significance and role as a nosocomial pathogen. J. Hosp. Infect. 2004, 57, 1–7. [Google Scholar] [CrossRef]
  194. Sader, H.S.; Jones, R.N. Antimicrobial susceptibility of uncommonly isolated non-enteric Gram-negative bacilli. Int. J. Antimicrob. Agents 2005, 25, 95–109. [Google Scholar] [CrossRef]
  195. Crossman, L.C.; Gould, V.C.; Dow, J.M.; Vernikos, G.S.; Okazaki, A.; Sebaihia, M.; Saunders, D.; Arrowsmith, C.; Carver, T.; Peters, N.; et al. The complete genome, comparative and functional analysis of Stenotrophomonas maltophilia reveals an organism heavily shielded by drug resistance determinants. Genome Biol. 2008, 9, R74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Denton, M.; Kerr, K.G. Microbiological and clinical aspects of infection associated with Stenotrophomonas maltophilia. Clin. Microbiol. Rev. 1998, 11, 57–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Nicodemo, A.C.; Araujo, M.R.; Ruiz, A.S.; Gales, A.C. In vitro susceptibility of Stenotrophomonas maltophilia isolates: Comparison of disc diffusion, Etest and agar dilution methods. J. Antimicrob. Chemother. 2004, 53, 604–608. [Google Scholar] [CrossRef] [Green Version]
  198. Li, X.Z.; Zhang, L.; McKay, G.A.; Poole, K. Role of the acetyltransferase AAC(6’)-Iz modifying enzyme in aminoglycoside resistance in Stenotrophomonas maltophilia. J. Antimicrob. Chemother. 2003, 51, 803–811. [Google Scholar] [CrossRef] [Green Version]
  199. Toleman, M.A.; Bennett, P.M.; Bennett, D.M.; Jones, R.N.; Walsh, T.R. Global emergence of trimethoprim/sulfamethoxazole resistance in Stenotrophomonas maltophilia mediated by acquisition of sul genes. Emerg. Infect. Dis. 2007, 13, 559–565. [Google Scholar] [CrossRef]
  200. Zelenitsky, S.A.; Iacovides, H.; Ariano, R.E.; Harding, G.K. Antibiotic combinations significantly more active than monotherapy in an in vitro infection model of Stenotrophomonas maltophilia. Diagn. Microbiol. Infect. Dis. 2005, 51, 39–43. [Google Scholar] [CrossRef]
  201. Cho, S.Y.; Kang, C.I.; Kim, J.; Ha, Y.E.; Chung, D.R.; Lee, N.Y.; Peck, K.R.; Song, J.H. Can levofloxacin be a useful alternative to trimethoprim-sulfamethoxazole for treating Stenotrophomonas maltophilia bacteremia? Antimicrob. Agents Chemother. 2014, 58, 581–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Biagi, M.; Tan, X.; Wu, T.; Jurkovic, M.; Vialichka, A.; Meyer, K.; Mendes, R.E.; Wenzler, E. Activity of Potential Alternative Treatment Agents for Stenotrophomonas maltophilia Isolates Nonsusceptible to Levofloxacin and/or Trimethoprim-Sulfamethoxazole. J. Clin. Microbiol. 2020, 58, e01603-19. [Google Scholar] [CrossRef]
  203. Ciacci, N.; Boncompagni, S.; Valzano, F.; Cariani, L.; Aliberti, S.; Blasi, F.; Pollini, S.; Rossolini, G.M.; Pallecchi, L. In Vitro Synergism of Colistin and N-acetylcysteine against Stenotrophomonas maltophilia. Antibiotics 2019, 8, 101. [Google Scholar] [CrossRef] [Green Version]
  204. Lira, F.; Hernandez, A.; Belda, E.; Sanchez, M.B.; Moya, A.; Silva, F.J.; Martinez, J.L. Whole-genome sequence of Stenotrophomonas maltophilia D457, a clinical isolate and a model strain. J. Bacteriol. 2012, 194, 3563–3564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Sanchez, M.B. Antibiotic resistance in the opportunistic pathogen Stenotrophomonas maltophilia. Front. Microbiol. 2015, 6, 658. [Google Scholar] [CrossRef] [Green Version]
  206. Lecso-Bornet, M.; Pierre, J.; Sarkis-Karam, D.; Lubera, S.; Bergogne-Berezin, E. Susceptibility of Xanthomonas maltophilia to six quinolones and study of outer membrane proteins in resistant mutants selected in vitro. Antimicrob. Agents Chemother. 1992, 36, 669–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Alonso, A.; Martinez, J.L. Cloning and characterization of SmeDEF, a novel multidrug efflux pump from Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2000, 44, 3079–3086. [Google Scholar] [CrossRef] [Green Version]
  208. Crowder, M.W.; Walsh, T.R.; Banovic, L.; Pettit, M.; Spencer, J. Overexpression, purification, and characterization of the cloned metallo-beta-lactamase L1 from Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 1998, 42, 921–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Walsh, T.R.; MacGowan, A.P.; Bennett, P.M. Sequence analysis and enzyme kinetics of the L2 serine beta-lactamase from Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 1997, 41, 1460–1464. [Google Scholar] [CrossRef] [Green Version]
  210. Hu, R.M.; Huang, K.J.; Wu, L.T.; Hsiao, Y.J.; Yang, T.C. Induction of L1 and L2 beta-lactamases of Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2008, 52, 1198–1200. [Google Scholar] [CrossRef] [Green Version]
  211. Okazaki, A.; Avison, M.B. Induction of L1 and L2 beta-lactamase production in Stenotrophomonas maltophilia is dependent on an AmpR-type regulator. Antimicrob. Agents Chemother. 2008, 52, 1525–1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Lin, C.W.; Huang, Y.W.; Hu, R.M.; Chiang, K.H.; Yang, T.C. The role of AmpR in regulation of L1 and L2 beta-lactamases in Stenotrophomonas maltophilia. Res. Microbiol. 2009, 160, 152–158. [Google Scholar] [CrossRef] [PubMed]
  213. Tada, T.; Miyoshi-Akiyama, T.; Dahal, R.K.; Mishra, S.K.; Shimada, K.; Ohara, H.; Kirikae, T.; Pokhrel, B.M. Identification of a novel 6′-N-aminoglycoside acetyltransferase, AAC(6′)-Iak, from a multidrug-resistant clinical isolate of Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2014, 58, 6324–6327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Lambert, T.; Ploy, M.C.; Denis, F.; Courvalin, P. Characterization of the chromosomal aac(6′)-Iz gene of Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 1999, 43, 2366–2371. [Google Scholar] [CrossRef] [Green Version]
  215. Okazaki, A.; Avison, M.B. Aph(3′)-IIc, an aminoglycoside resistance determinant from Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2007, 51, 359–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Sanchez, M.B.; Hernandez, A.; Rodriguez-Martinez, J.M.; Martinez-Martinez, L.; Martinez, J.L. Predictive analysis of transmissible quinolone resistance indicates Stenotrophomonas maltophilia as a potential source of a novel family of Qnr determinants. BMC Microbiol. 2008, 8, 148. [Google Scholar] [CrossRef] [Green Version]
  217. Shimizu, K.; Kikuchi, K.; Sasaki, T.; Takahashi, N.; Ohtsuka, M.; Ono, Y.; Hiramatsu, K. Smqnr, a new chromosome-carried quinolone resistance gene in Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2008, 52, 3823–3825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Sanchez, M.B.; Martinez, J.L. SmQnr contributes to intrinsic resistance to quinolones in Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2010, 54, 580–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Gordon, N.C.; Wareham, D.W. Novel variants of the Smqnr family of quinolone resistance genes in clinical isolates of Stenotrophomonas maltophilia. J. Antimicrob. Chemother. 2010, 65, 483–489. [Google Scholar] [CrossRef]
  220. Lin, Y.T.; Huang, Y.W.; Chen, S.J.; Chang, C.W.; Yang, T.C. The SmeYZ Efflux Pump of Stenotrophomonas maltophilia Contributes to Drug Resistance, Virulence-Related Characteristics, and Virulence in Mice. Antimicrob. Agents Chemother. 2015, 59, 4067–4073. [Google Scholar] [CrossRef] [Green Version]
  221. Blanco, P.; Corona, F.; Martinez, J.L. Involvement of the RND efflux pump transporter SmeH in the acquisition of resistance to ceftazidime in Stenotrophomonas maltophilia. Sci. Rep. 2019, 9, 4917. [Google Scholar] [CrossRef]
  222. Gould, V.C.; Okazaki, A.; Avison, M.B. Coordinate hyperproduction of SmeZ and SmeJK efflux pumps extends drug resistance in Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2013, 57, 655–657. [Google Scholar] [CrossRef] [Green Version]
  223. Lin, C.W.; Huang, Y.W.; Hu, R.M.; Yang, T.C. SmeOP-TolCSm efflux pump contributes to the multidrug resistance of Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2014, 58, 2405–2408. [Google Scholar] [CrossRef] [Green Version]
  224. Sanchez, P.; Alonso, A.; Martinez, J.L. Cloning and characterization of SmeT, a repressor of the Stenotrophomonas maltophilia multidrug efflux pump SmeDEF. Antimicrob. Agents Chemother. 2002, 46, 3386–3393. [Google Scholar] [CrossRef] [Green Version]
  225. Sanchez, M.B.; Martinez, J.L. Overexpression of the Efflux Pumps SmeVWX and SmeDEF Is a Major Cause of Resistance to Co-trimoxazole in Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2018, 62, e00301-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Wu, C.J.; Lu, H.F.; Lin, Y.T.; Zhang, M.S.; Li, L.H.; Yang, T.C. Substantial Contribution of SmeDEF, SmeVWX, SmQnr, and Heat Shock Response to Fluoroquinolone Resistance in Clinical Isolates of Stenotrophomonas maltophilia. Front. Microbiol. 2019, 10, 822. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, L.; Li, X.Z.; Poole, K. SmeDEF multidrug efflux pump contributes to intrinsic multidrug resistance in Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2001, 45, 3497–3503. [Google Scholar] [CrossRef] [Green Version]
  228. Sanchez, P.; Moreno, E.; Martinez, J.L. The biocide triclosan selects Stenotrophomonas maltophilia mutants that overproduce the SmeDEF multidrug efflux pump. Antimicrob. Agents Chemother. 2005, 49, 781–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Hernandez, A.; Ruiz, F.M.; Romero, A.; Martinez, J.L. The binding of triclosan to SmeT, the repressor of the multidrug efflux pump SmeDEF, induces antibiotic resistance in Stenotrophomonas maltophilia. PLoS Pathog. 2011, 7, e1002103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Al-Hamad, A.; Upton, M.; Burnie, J. Molecular cloning and characterization of SmrA, a novel ABC multidrug efflux pump from Stenotrophomonas maltophilia. J. Antimicrob. Chemother. 2009, 64, 731–734. [Google Scholar] [CrossRef] [Green Version]
  231. Lin, Y.T.; Huang, Y.W.; Liou, R.S.; Chang, Y.C.; Yang, T.C. MacABCsm, an ABC-type tripartite efflux pump of Stenotrophomonas maltophilia involved in drug resistance, oxidative and envelope stress tolerances and biofilm formation. J. Antimicrob. Chemother. 2014, 69, 3221–3226. [Google Scholar] [CrossRef]
  232. Huang, Y.W.; Hu, R.M.; Chu, F.Y.; Lin, H.R.; Yang, T.C. Characterization of a major facilitator superfamily (MFS) tripartite efflux pump EmrCABsm from Stenotrophomonas maltophilia. J. Antimicrob. Chemother. 2013, 68, 2498–2505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Mett, H.; Rosta, S.; Schacher, B.; Frei, R. Outer membrane permeability and beta-lactamase content in Pseudomonas maltophilia clinical isolates and laboratory mutants. Rev. Infect. Dis. 1988, 10, 765–769. [Google Scholar] [CrossRef]
  234. Gil-Gil, T.; Martinez, J.L.; Blanco, P. Mechanisms of antimicrobial resistance in Stenotrophomonas maltophilia: A review of current knowledge. Expert Rev. Anti. Infect. Ther. 2020, 18, 335–347. [Google Scholar] [CrossRef]
  235. Valdezate, S.; Vindel, A.; Echeita, A.; Baquero, F.; Canton, R. Topoisomerase II and IV quinolone resistance-determining regions in Stenotrophomonas maltophilia clinical isolates with different levels of quinolone susceptibility. Antimicrob. Agents Chemother. 2002, 46, 665–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Ribera, A.; Domenech-Sanchez, A.; Ruiz, J.; Benedi, V.J.; Jimenez de Anta, M.T.; Vila, J. Mutations in gyrA and parC QRDRs are not relevant for quinolone resistance in epidemiological unrelated Stenotrophomonas maltophilia clinical isolates. Microb. Drug Resist. 2002, 8, 245–251. [Google Scholar] [CrossRef]
  237. Blair, J.M.; Bavro, V.N.; Ricci, V.; Modi, N.; Cacciotto, P.; Kleinekathfer, U.; Ruggerone, P.; Vargiu, A.V.; Baylay, A.J.; Smith, H.E.; et al. AcrB drug-binding pocket substitution confers clinically relevant resistance and altered substrate specificity. Proc. Natl. Acad. Sci. USA 2015, 112, 3511–3516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Chang, L.L.; Chen, H.F.; Chang, C.Y.; Lee, T.M.; Wu, W.J. Contribution of integrons, and SmeABC and SmeDEF efflux pumps to multidrug resistance in clinical isolates of Stenotrophomonas maltophilia. J. Antimicrob. Chemother. 2004, 53, 518–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Gil-Gil, T.; Corona, F.; Martínez, J.L.; Bernardini, A. The Inactivation of Enzymes Belonging to the Central Carbon Metabolism Is a Novel Mechanism of Developing Antibiotic Resistance. Msystems 2020, 5, e00282-20. [Google Scholar] [CrossRef] [PubMed]
  240. Kanamori, H.; Yano, H.; Tanouchi, A.; Kakuta, R.; Endo, S.; Ichimura, S.; Ogawa, M.; Shimojima, M.; Inomata, S.; Ozawa, D.; et al. Prevalence of Smqnr and plasmid-mediated quinolone resistance determinants in clinical isolates of Stenotrophomonas maltophilia from Japan: Novel variants of Smqnr. New Microbes New Infect. 2015, 7, 8–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Furlan, J.P.R.; Sanchez, D.G.; Gallo, I.F.L.; Stehling, E.G. Characterization of Acquired Antimicrobial Resistance Genes in Environmental Stenotrophomonas maltophilia Isolates from Brazil. Microb. Drug Resist. 2019, 25, 475–479. [Google Scholar] [CrossRef]
  242. Peters, D.L.; McCutcheon, J.G.; Stothard, P.; Dennis, J.J. Novel Stenotrophomonas maltophilia temperate phage DLP4 is capable of lysogenic conversion. BMC Genom. 2019, 20, 300. [Google Scholar] [CrossRef] [PubMed]
  243. Rahmati-Bahram, A.; Magee, J.T.; Jackson, S.K. Temperature-dependent aminoglycoside resistance in Stenotrophomonas (Xanthomonas) maltophilia; alterations in protein and lipopolysaccharide with growth temperature. J. Antimicrob. Chemother. 1996, 37, 665–676. [Google Scholar] [CrossRef] [Green Version]
  244. Hall, C.W.; Mah, T.F. Molecular mechanisms of biofilm-based antibiotic resistance and tolerance in pathogenic bacteria. FEMS Microbiol. Rev. 2017, 41, 276–301. [Google Scholar] [CrossRef]
  245. Ryan, R.P.; Fouhy, Y.; Garcia, B.F.; Watt, S.A.; Niehaus, K.; Yang, L.; Tolker-Nielsen, T.; Dow, J.M. Interspecies signalling via the Stenotrophomonas maltophilia diffusible signal factor influences biofilm formation and polymyxin tolerance in Pseudomonas aeruginosa. Mol. Microbiol. 2008, 68, 75–86. [Google Scholar] [CrossRef] [PubMed]
  246. Garcia-Leon, G.; Hernandez, A.; Hernando-Amado, S.; Alavi, P.; Berg, G.; Martinez, J.L. A function of SmeDEF, the major quinolone resistance determinant of Stenotrophomonas maltophilia, is the colonization of plant roots. Appl. Environ. Microbiol. 2014, 80, 4559–4565. [Google Scholar] [CrossRef] [Green Version]
  247. Blanco, P.; Corona, F.; Martinez, J.L. Biolog Phenotype Microarray Is a Tool for the Identification of Multidrug Resistance Efflux Pump Inducers. Antimicrob. Agents Chemother. 2018, 62, e01263-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Blanco, P.; Corona, F.; Sanchez, M.B.; Martinez, J.L. Vitamin K3 Induces the Expression of the Stenotrophomonas maltophilia SmeVWX Multidrug Efflux Pump. Antimicrob. Agents Chemother. 2017, 61, e02453-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Hu, R.M.; Liao, S.T.; Huang, C.C.; Huang, Y.W.; Yang, T.C. An inducible fusaric acid tripartite efflux pump contributes to the fusaric acid resistance in Stenotrophomonas maltophilia. PLoS ONE 2012, 7, e51053. [Google Scholar]
  250. Depoorter, E.; De Canck, E.; Peeters, C.; Wieme, A.D.; Cnockaert, M.; Zlosnik, J.E.A.; LiPuma, J.J.; Coenye, T.; Vandamme, P. Burkholderia cepacia Complex Taxon K: Where to Split? Front. Microbiol. 2020, 11, 1594. [Google Scholar] [CrossRef]
  251. Depoorter, E.; Bull, M.J.; Peeters, C.; Coenye, T.; Vandamme, P.; Mahenthiralingam, E. Burkholderia: An update on taxonomy and biotechnological potential as antibiotic producers. Appl. Microbiol. Biotechnol. 2016, 100, 5215–5229. [Google Scholar] [CrossRef] [PubMed]
  252. Coenye, T.; Mahenthiralingam, E.; Henry, D.; LiPuma, J.J.; Laevens, S.; Gillis, M.; Speert, D.P.; Vandamme, P. Burkholderia ambifaria sp. nov., a novel member of the Burkholderia cepacia complex including biocontrol and cystic fibrosis-related isolates. Int. J. Syst. Evol. Microbiol. 2001, 51, 1481–1490. [Google Scholar] [CrossRef] [Green Version]
  253. Parke, J.L.; Gurian-Sherman, D. Diversity of the Burkholderia cepacia C complex and implications for rsk assessment of biological control strains Annu. Rev. Phytopathol. 2001, 39, 225–258. [Google Scholar] [CrossRef]
  254. Kunakom, S.; Eustáquio, A.S. Burkholderia as a Source of Natural Products. J. Nat. Prod. 2019, 82, 2018–2037. [Google Scholar] [CrossRef] [PubMed]
  255. Coenye, T.; LiPuma, J.J. Multilocus Restriction Typing: A Novel Tool for Studying Global Epidemiology of Burkholderia cepacia Complex Infection in Cystic Fibrosis. J. Infect. Dis. 2002, 185, 1454–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Mahenthiralingam, E.; Baldwin, A.; Dowson, C.G. Burkholderia cepacia complex bacteria: Opportunistic pathogens with important natural biology. J. Appl. Microbiol. 2008, 104, 1539–1551. [Google Scholar] [CrossRef]
  257. Sousa, S.A.; Ramos, C.G.; Leitão, J.H. Burkholderia cepacia Complex: Emerging Multihost Pathogens Equipped with a Wide Range of Virulence Factors and Determinants. Int. J. Microbiol. 2011, 2011, 1–9. [Google Scholar] [CrossRef] [Green Version]
  258. Dobbin, C.J.; Soni, R.; Jelihovsky, T.; Bye, P.T. Cepacia syndrome occurring following prolonged colonisation with Burkholderia cepacia. Aust. N. Z. J. Med. 2000, 30, 288–289. [Google Scholar] [CrossRef]
  259. Somayaji, R.; Yau, Y.C.W.; Tullis, E.; LiPuma, J.J.; Ratjen, F.; Waters, V. Clinical Outcomes Associated with Burkholderia cepacia Complex Infection in Patients with Cystic Fibrosis. Ann. Am. Thorac. Soc. 2020, 17, 1542–1548. [Google Scholar] [CrossRef]
  260. Husain, S.; Singh, N. Burkholderia cepacia infection and lung transplantation. Semin. Respir. Infect. 2002, 17, 284–290. [Google Scholar] [CrossRef] [PubMed]
  261. Reik, R.; Spilker, T.; LiPuma, J.J. Distribution of Burkholderia cepacia Complex Species among Isolates Recovered from Persons with or without Cystic Fibrosis. J. Clin. Microbiol. 2005, 43, 2926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Kenna, D.T.D.; Lilley, D.; Coward, A.; Martin, K.; Perry, C.; Pike, R.; Hill, R.; Turton, J.F. Prevalence of Burkholderia species, including members of Burkholderia cepacia complex, among UK cystic and non-cystic fibrosis patients. J. Med Microbiol. 2017, 66, 490–501. [Google Scholar] [CrossRef] [PubMed]
  263. Power, R.F.; Linnane, B.; Martin, R.; Power, N.; Harnett, P.; Casserly, B.; O’Connell, N.H.; Dunne, C.P. The first reported case of Burkholderia contaminans in patients with cystic fibrosis in Ireland: From the Sargasso Sea to Irish Children. BMC Pulm. Med. 2016, 16, 57. [Google Scholar] [CrossRef] [Green Version]
  264. Zurita, J.; Mejia, L.; Zapata, S.; Trueba, G.; Vargas, A.C.; Aguirre, S.; Falconi, G. Healthcare-associated respiratory tract infection and colonization in an intensive care unit caused by Burkholderia cepacia isolated in mouthwash. Int. J. Infect. Dis. 2014, 29, 96–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Souza, A.V.; Moreira, C.R.; Pasternak, J.; Hirata, M.d.L.; Saltini, D.A.; Caetano, V.C.; Ciosak, S.; Azevedo, F.M.; Severino, P.; Vandamme, P.; et al. Characterizing uncommon Burkholderia cepacia complex isolates from an outbreak in a haemodialysis unit. J. Med Microbiol. 2004, 53, 999–1005. [Google Scholar] [CrossRef] [Green Version]
  266. Balandreau, J.; Viallard, V.; Cournoyer, B.; Coenye, T.; Laevens, S.; Vandamme, P. Burkholderia cepacia Genomovar III Is a Common Plant-Associated Bacterium. Appl. Environ. Microbiol. 2001, 67, 982–985. [Google Scholar] [CrossRef] [Green Version]
  267. Fiore, A.; Laevens, S.; Bevivino, A.; Dalmastri, C.; Tabacchioni, S.; Vandamme, P.; Chiarini, L. Burkholderia cepacia complex: Distribution of genomovars among isolates from the maize rhizosphere in Italy. Environ. Microbiol. 2001, 3, 137–143. [Google Scholar] [CrossRef] [PubMed]
  268. Baldwin, A.; Mahenthiralingam, E.; Drevinek, P.; Vandamme, P.; Govan, J.R.; Waine, D.J.; LiPuma, J.J.; Chiarini, L.; Dalmastri, C.; Henry, D.A.; et al. Environmental Burkholderia cepacia complex isolates in human infections. Emerg. Infect. Dis. 2007, 13, 458–461. [Google Scholar] [CrossRef]
  269. Springman, A.C.; Jacobs, J.L.; Somvanshi, V.S.; Sundin, G.W.; Mulks, M.H.; Whittam, T.S.; Viswanathan, P.; Gray, R.L.; Lipuma, J.J.; Ciche, T.A. Genetic diversity and multihost pathogenicity of clinical and environmental strains of Burkholderia cenocepacia. Appl. Environ. Microbiol. 2009, 75, 5250–5260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  270. Coenye, T. Population structure analysis of Burkholderia cepacia genomovar III: Varying degrees of genetic recombination characterize major clonal complexes. Microbiology 2003, 149, 77–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Nzula, S.; Vandamme, P.; Govan, J.R. Influence of taxonomic status on the in vitro antimicrobial susceptibility of the Burkholderia cepacia complex. J. Antimicrob. Chemother. 2002, 50, 265–269. [Google Scholar] [CrossRef] [PubMed]
  272. Nummila, K.; Kilpeläinen, I.; Zähringer, U.; Vaara, M.; Helander, I.M. Lipopolysaccharides of polymyxin B-resistant mutants of Escherichia coli are extensively substituted by 2-aminoethyl pyrophosphate and contain aminoarabinose in lipid A. Mol. Microbiol. 1995, 16, 271–278. [Google Scholar] [CrossRef] [PubMed]
  273. Ernst, R.K.; Yi, E.C.; Guo, L.; Lim, K.B.; Burns, J.L.; Hackett, M.; Miller, S.I. Specific Lipopolysaccharide Found in Cystic Fibrosis Airway Pseudomonas aeruginosa. Science 1999, 286, 1561. [Google Scholar] [CrossRef] [PubMed]
  274. Ortega, X.P.; Cardona, S.T.; Brown, A.R.; Loutet, S.A.; Flannagan, R.S.; Campopiano, D.J.; Govan, J.R.; Valvano, M.A. A putative gene cluster for aminoarabinose biosynthesis is essential for Burkholderia cenocepacia viability. J. Bacteriol. 2007, 189, 3639–3644. [Google Scholar] [CrossRef] [Green Version]
  275. Poirel, L.; Rodriguez-Martinez, J.M.; Plésiat, P.; Nordmann, P. Naturally occurring Class A ss-lactamases from the Burkholderia cepacia complex. Antimicrob. Agents Chemother. 2009, 53, 876–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Trépanier, S.; Prince, A.; Huletsky, A. Characterization of the penA and penR genes of Burkholderia cepacia 249 which encode the chromosomal class A penicillinase and its LysR-type transcriptional regulator. Antimicrob. Agents Chemother. 1997, 41, 2399–2405. [Google Scholar] [CrossRef] [Green Version]
  277. Becka, S.A.; Zeiser, E.T.; Barnes, M.D.; Taracila, M.A.; Nguyen, K.; Singh, I.; Sutton, G.G.; LiPuma, J.J.; Fouts, D.E.; Papp-Wallace, K.M. Characterization of the AmpC β-Lactamase from Burkholderia multivorans. Antimicrob. Agents Chemother. 2018, 62, e01140-18. [Google Scholar] [CrossRef] [Green Version]
  278. Hwang, J.; Kim, H.S. Cell Wall Recycling-Linked Coregulation of AmpC and PenB β-Lactamases through ampD Mutations in Burkholderia cenocepacia. Antimicrob. Agents Chemother. 2015, 59, 7602–7610. [Google Scholar] [CrossRef] [Green Version]
  279. Nukaga, M.; Yoon, M.J.; Taracilia, M.A.; Hoshino, T.; Becka, S.A.; Zeiser, E.T.; Johnson, J.R.; Papp-Wallace, K.M. Assessing the Potency of β-Lactamase Inhibitors with Diverse Inactivation Mechanisms against the PenA1 Carbapenemase from Burkholderia multivorans. ACS Infect. Dis. 2021, 7, 826–837. [Google Scholar] [CrossRef]
  280. Fehlner-Gardiner, C.C.; Valvano, M.A. Cloning and characterization of the Burkholderia vietnamiensis norM gene encoding a multi-drug efflux protein. FEMS Microbiol. Lett. 2002, 215, 279–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  281. Guglierame, P.; Pasca, M.R.; De Rossi, E.; Buroni, S.; Arrigo, P.; Manina, G.; Riccardi, G. Efflux pump genes of the resistance-nodulation-division family in Burkholderia cenocepacia genome. BMC Microbiol. 2006, 6, 66. [Google Scholar] [CrossRef] [Green Version]
  282. Podnecky, N.L.; Rhodes, K.A.; Schweizer, H.P. Efflux pump-mediated drug resistance in Burkholderia. Front. Microbiol. 2015, 6, 305. [Google Scholar] [CrossRef] [Green Version]
  283. Buroni, S.; Pasca, M.R.; Flannagan, R.S.; Bazzini, S.; Milano, A.; Bertani, I.; Venturi, V.; Valvano, M.A.; Riccardi, G. Assessment of three Resistance-Nodulation-Cell Division drug efflux transporters of Burkholderia cenocepacia in intrinsic antibiotic resistance. BMC Microbiol. 2009, 9, 200. [Google Scholar] [CrossRef] [PubMed]
  284. Wigfield, S.M.; Rigg, G.P.; Kavari, M.; Webb, A.K.; Matthews, R.C.; Burnie, J.P. Identification of an immunodominant drug efflux pump in Burkholderia cepacia. J. Antimicrob. Chemother. 2002, 49, 619–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Nair, B.M.; Cheung, K.J., Jr.; Griffith, A.; Burns, J.L. Salicylate induces an antibiotic efflux pump in Burkholderia cepacia complex genomovar III (B. cenocepacia). J. Clin. Investig. 2004, 113, 464–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. El-Halfawy, O.M.; Klett, J.; Ingram, R.J.; Loutet, S.A.; Murphy, M.E.; Martín-Santamaría, S.; Valvano, M.A. Antibiotic Capture by Bacterial Lipocalins Uncovers an Extracellular Mechanism of Intrinsic Antibiotic Resistance. Mbio 2017, 8, e00225-17. [Google Scholar] [CrossRef] [Green Version]
  287. Diaz Caballero, J.; Clark, S.T.; Wang, P.W.; Donaldson, S.L.; Coburn, B.; Tullis, D.E.; Yau, Y.C.W.; Waters, V.J.; Hwang, D.M.; Guttman, D.S. A genome-wide association analysis reveals a potential role for recombination in the evolution of antimicrobial resistance in Burkholderia multivorans. Plos Pathog. 2018, 14, e1007453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Tseng, S.P.; Tsai, W.C.; Liang, C.Y.; Lin, Y.S.; Huang, J.W.; Chang, C.Y.; Tyan, Y.C.; Lu, P.L. The contribution of antibiotic resistance mechanisms in clinical Burkholderia cepacia complex isolates: An emphasis on efflux pump activity. PLoS ONE 2014, 9, e104986. [Google Scholar] [CrossRef]
  289. Lennon, E.; DeCicco, B.T. Plasmids of Pseudomonas cepacia strains of diverse origins. Appl. Environ. Microbiol. 1991, 57, 2345–2350. [Google Scholar] [CrossRef] [Green Version]
  290. Roszniowski, B.; McClean, S.; Drulis-Kawa, Z. Burkholderia cenocepacia Prophages-Prevalence, Chromosome Location and Major Genes Involved. Viruses 2018, 10, 297. [Google Scholar] [CrossRef] [Green Version]
  291. Vouga, M.; Greub, G. Emerging bacterial pathogens: The past and beyond. Clin. Microbiol. Infect. 2016, 22, 12–21. [Google Scholar] [CrossRef] [Green Version]
  292. Davies, J.; Davies, D. Origins and evolution of antibiotic resistance. Microbiol. Mol. Biol. Rev. 2010, 74, 417–433. [Google Scholar] [CrossRef] [Green Version]
  293. Martinez, J.L. Ecology and Evolution of Chromosomal Gene Transfer between Environmental Microorganisms and Pathogens. Microbiol. Spectr. 2018, 6. [Google Scholar] [CrossRef]
  294. Ryan, M.P.; Pembroke, J.T. Brevundimonas spp: Emerging global opportunistic pathogens. Virulence 2018, 9, 480–493. [Google Scholar] [CrossRef] [Green Version]
  295. Segers, P.; Vancanneyt, M.; Pot, B.; Torck, U.; Hoste, B.; Dewettinck, D.; Falsen, E.; Kersters, K.; De Vos, P. Classification of Pseudomonas diminuta Leifson and Hugh 1954 and Pseudomonas vesicularis Busing, Doll, and Freytag 1953 in Brevundimonas gen. nov. as Brevundimonas diminuta comb. nov. and Brevundimonas vesicularis comb. nov., respectively. Int. J. Syst. Bacteriol. 1994, 44, 499–510. [Google Scholar] [CrossRef]
  296. Menuet, M.; Bittar, F.; Stremler, N.; Dubus, J.C.; Sarles, J.; Raoult, D.; Rolain, J.M. First isolation of two colistin-resistant emerging pathogens, Brevundimonas diminuta and Ochrobactrum anthropi, in a woman with cystic fibrosis: A case report. J. Med. Case Rep. 2008, 2, 373. [Google Scholar] [CrossRef] [PubMed]
  297. Karadag, N.; Karagol, B.S.; Kundak, A.A.; Dursun, A.; Okumus, N.; Tanir, G.; Zenciroglu, A. Spectrum of Brevundimonas vesicularis infections in neonatal period: A case series at a tertiary referral center. Infection 2012, 40, 509–515. [Google Scholar] [CrossRef]
  298. Fernandez-Olmos, A.; Garcia-Castillo, M.; Morosini, M.I.; Lamas, A.; Maiz, L.; Canton, R. MALDI-TOF MS improves routine identification of non-fermenting Gram negative isolates from cystic fibrosis patients. J. Cyst. Fibros. 2012, 11, 59–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  299. Zhang, C.C.; Hsu, H.J.; Li, C.M. Brevundimonas vesicularis bacteremia resistant to trimethoprim-sulfamethoxazole and ceftazidime in a tertiary hospital in southern Taiwan. J. Microbiol. Immunol. Infect. 2012, 45, 448–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  300. Gilad, J.; Borer, A.; Peled, N.; Riesenberg, K.; Tager, S.; Appelbaum, A.; Schlaeffer, F. Hospital-acquired brevundimonas vesicularis septicaemia following open-heart surgery: Case report and literature review. Scand. J. Infect. Dis. 2000, 32, 90–91. [Google Scholar]
  301. Han, X.Y.; Andrade, R.A. Brevundimonas diminuta infections and its resistance to fluoroquinolones. J. Antimicrob. Chemother. 2005, 55, 853–859. [Google Scholar] [CrossRef] [Green Version]
  302. Almuzara, M.N.; Barberis, C.M.; Rodriguez, C.H.; Famiglietti, A.M.; Ramirez, M.S.; Vay, C.A. First report of an extensively drug-resistant VIM-2 metallo-beta-lactamase-producing Brevundimonas diminuta clinical isolate. J. Clin. Microbiol. 2012, 50, 2830–2832. [Google Scholar] [CrossRef] [Green Version]
  303. Adelowo, O.O.; Fagade, O.E. The tetracycline resistance gene tet39 is present in both Gram-negative and Gram-positive bacteria from a polluted river, Southwestern Nigeria. Lett. Appl. Microbiol. 2009, 48, 167–172. [Google Scholar] [CrossRef]
  304. Guermouche M’rassi, A.; Bensalah, F.; Gury, J.; Duran, R. Isolation and characterization of different bacterial strains for bioremediation of n-alkanes and polycyclic aromatic hydrocarbons. Environ. Sci. Pollut. Res. Int. 2015, 22, 15332–15346. [Google Scholar] [CrossRef]
  305. Simidu, U.; Kita-Tsukamoto, K.; Yasumoto, T.; Yotsu, M. Taxonomy of four marine bacterial strains that produce tetrodotoxin. Int. J. Syst. Bacteriol. 1990, 40, 331–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Brink, A.J.; van Straten, A.; van Rensburg, A.J. Shewanella (Pseudomonas) putrefaciens bacteremia. Clin. Infect. Dis. 1995, 20, 1327–1332. [Google Scholar] [CrossRef] [PubMed]
  307. Jacob-Kokura, S.; Chan, C.Y.; Kaplan, L. Bacteremia and empyema caused by Shewanella algae in a trauma patient. Ann. Pharmacother. 2014, 48, 128–136. [Google Scholar] [CrossRef] [PubMed]
  308. Appelbaum, P.C.; Bowen, A.J. Opportunistic infection of chronic skin ulcers with Pseudomonas putrefaciens. Br. J. Dermatol. 1978, 98, 229–231. [Google Scholar] [CrossRef] [PubMed]
  309. Debois, J.; Degreef, H.; Vandepitte, J.; Spaepen, J. Pseudomonas putrefaciens as a cause of infection in humans. J. Clin. Pathol. 1975, 28, 993–996. [Google Scholar] [CrossRef] [Green Version]
  310. Holmes, B.; Lapage, S.P.; Malnick, H. Strains of Pseudomonas putrefaciens from clinical material. J. Clin. Pathol. 1975, 28, 149–155. [Google Scholar] [CrossRef]
  311. Holt, H.M.; Sogaard, P.; Gahrn-Hansen, B. Ear infections with Shewanella alga: A bacteriologic, clinical and epidemiologic study of 67 cases. Clin. Microbiol. Infect. 1997, 3, 329–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Vogel, B.F.; Holt, H.M.; Gerner-Smidt, P.; Bundvad, A.; Sogaard, P.; Gram, L. Homogeneity of Danish environmental and clinical isolates of Shewanella algae. Appl. Environ. Microbiol. 2000, 66, 443–448. [Google Scholar] [CrossRef] [Green Version]
  313. Nozue, H.; Hayashi, T.; Hashimoto, Y.; Ezaki, T.; Hamasaki, K.; Ohwada, K.; Terawaki, Y. Isolation and characterization of Shewanella alga from human clinical specimens and emendation of the description of S. alga Simidu et al., 1990, 335. Int. J. Syst. Bacteriol. 1992, 42, 628–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Janda, J.M.; Abbott, S.L. The genus Shewanella: From the briny depths below to human pathogen. Crit. Rev. Microbiol. 2014, 40, 293–312. [Google Scholar] [CrossRef] [PubMed]
  315. Zago, V.; Veschetti, L.; Patuzzo, C.; Malerba, G.; Lleo, M.M. Shewanella algae and Vibrio spp. strains isolated in Italian aquaculture farms are reservoirs of antibiotic resistant genes that might constitute a risk for human health. Mar. Pollut. Bull. 2020, 154, 111057. [Google Scholar] [CrossRef]
  316. Zago, V.; Veschetti, L.; Patuzzo, C.; Malerba, G.; Lleo, M.M. Resistome, Mobilome and Virulome Analysis of Shewanella algae and Vibrio spp. Strains Isolated in Italian Aquaculture Centers. Microorganisms 2020, 8, 572. [Google Scholar] [CrossRef] [Green Version]
  317. Poirel, L.; Rodriguez-Martinez, J.M.; Mammeri, H.; Liard, A.; Nordmann, P. Origin of plasmid-mediated quinolone resistance determinant QnrA. Antimicrob. Agents Chemother. 2005, 49, 3523–3525. [Google Scholar] [CrossRef] [Green Version]
  318. Potron, A.; Poirel, L.; Nordmann, P. Origin of OXA-181, an emerging carbapenem-hydrolyzing oxacillinase, as a chromosomal gene in Shewanella xiamenensis. Antimicrob. Agents Chemother. 2011, 55, 4405–4407. [Google Scholar] [CrossRef] [Green Version]
  319. Yousfi, K.; Bekal, S.; Usongo, V.; Touati, A. Current trends of human infections and antibiotic resistance of the genus Shewanella. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2017, 36, 1353–1362. [Google Scholar] [CrossRef]
  320. Latif, A.; Kapoor, V.; Vivekanandan, R.; Reddy, J.T. A rare case of Shewanella septicemia: Risk factors, environmental associations and management. BMJ Case Rep. 2019, 12, e230252. [Google Scholar] [CrossRef]
  321. Hong, J.; Steen, C.; Wong, E.; Keong, B. Shewanella: An important, emerging and lethal pathogen in a patient with recurrent presentations of cholangitis. BMJ Case Rep. 2020, 13, e237655. [Google Scholar] [CrossRef]
  322. Li, X.; Hu, Y.; Gong, J.; Zhang, L.; Wang, G. Comparative genome characterization of Achromobacter members reveals potential genetic determinants facilitating the adaptation to a pathogenic lifestyle. Appl. Microbiol. Biotechnol. 2013, 97, 6413–6425. [Google Scholar] [CrossRef] [PubMed]
  323. Mandell, W.F.; Garvey, G.J.; Neu, H.C. Achromobacter xylosoxidans bacteremia. Rev. Infect. Dis 1987, 9, 1001–1005. [Google Scholar] [CrossRef] [PubMed]
  324. Behrens-Muller, B.; Conway, J.; Yoder, J.; Conover, C.S. Investigation and control of an outbreak of Achromobacter xylosoxidans bacteremia. Infect. Control Hosp. Epidemiol. 2012, 33, 180–184. [Google Scholar] [CrossRef]
  325. Gomez-Cerezo, J.; Suarez, I.; Rios, J.J.; Pena, P.; Garcia de Miguel, M.J.; de Jose, M.; Monteagudo, O.; Linares, P.; Barbado-Cano, A.; Vazquez, J.J. Achromobacter xylosoxidans bacteremia: A 10-year analysis of 54 cases. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2003, 22, 360–363. [Google Scholar] [CrossRef] [PubMed]
  326. Amoureux, L.; Bador, J.; Siebor, E.; Taillefumier, N.; Fanton, A.; Neuwirth, C. Epidemiology and resistance of Achromobacter xylosoxidans from cystic fibrosis patients in Dijon, Burgundy: First French data. J. Cyst. Fibros. 2013, 12, 170–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Amoureux, L.; Bador, J.; Bounoua Zouak, F.; Chapuis, A.; de Curraize, C.; Neuwirth, C. Distribution of the species of Achromobacter in a French Cystic Fibrosis Centre and multilocus sequence typing analysis reveal the predominance of A. xylosoxidans and clonal relationships between some clinical and environmental isolates. J. Cyst. Fibros. 2016, 15, 486–494. [Google Scholar] [CrossRef] [Green Version]
  328. Isler, B.; Kidd, T.J.; Stewart, A.G.; Harris, P.; Paterson, D.L. Achromobacter Infections and Treatment Options. Antimicrob. Agents Chemother. 2020, 64, e01025-20. [Google Scholar] [CrossRef]
  329. Ridderberg, W.; Wang, M.; Norskov-Lauritsen, N. Multilocus sequence analysis of isolates of Achromobacter from patients with cystic fibrosis reveals infecting species other than Achromobacter xylosoxidans. J. Clin. Microbiol. 2012, 50, 2688–2694. [Google Scholar] [CrossRef] [Green Version]
  330. Bador, J.; Amoureux, L.; Duez, J.M.; Drabowicz, A.; Siebor, E.; Llanes, C.; Neuwirth, C. First description of an RND-type multidrug efflux pump in Achromobacter xylosoxidans, AxyABM. Antimicrob. Agents Chemother. 2011, 55, 4912–4914. [Google Scholar] [CrossRef] [Green Version]
  331. Bador, J.; Amoureux, L.; Blanc, E.; Neuwirth, C. Innate aminoglycoside resistance of Achromobacter xylosoxidans is due to AxyXY-OprZ, an RND-type multidrug efflux pump. Antimicrob. Agents Chemother. 2013, 57, 603–605. [Google Scholar] [CrossRef] [Green Version]
  332. El Salabi, A.; Borra, P.S.; Toleman, M.A.; Samuelsen, O.; Walsh, T.R. Genetic and biochemical characterization of a novel metallo-beta-lactamase, TMB-1, from an Achromobacter xylosoxidans strain isolated in Tripoli, Libya. Antimicrob. Agents Chemother. 2012, 56, 2241–2245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Yamamoto, M.; Nagao, M.; Hotta, G.; Matsumura, Y.; Matsushima, A.; Ito, Y.; Takakura, S.; Ichiyama, S. Molecular characterization of IMP-type metallo-beta-lactamases among multidrug-resistant Achromobacter xylosoxidans. J. Antimicrob. Chemother. 2012, 67, 2110–2113. [Google Scholar] [CrossRef] [Green Version]
  334. Gabrielaite, M.; Bartell, J.A.; Norskov-Lauritsen, N.; Pressler, T.; Nielsen, F.C.; Johansen, H.K.; Marvig, R.L. Transmission and antibiotic resistance of Achromobacter in cystic fibrosis. J. Clin. Microbiol. 2021, 59, e02911-20. [Google Scholar] [CrossRef] [PubMed]
  335. Chen, C.Y.; Hansen, K.S.; Hansen, L.K. Rhizobium radiobacter as an opportunistic pathogen in central venous catheter-associated bloodstream infection: Case report and review. J. Hosp. Infect. 2008, 68, 203–207. [Google Scholar] [CrossRef] [PubMed]
  336. Plotkin, G.R. Agrobacterium radiobacter prosthetic valve endocarditis. Ann. Intern. Med. 1980, 93, 839–840. [Google Scholar] [CrossRef] [PubMed]
  337. Paphitou, N.I.; Rolston, K.V. Catheter-related bacteremia caused by Agrobacterium radiobacter in a cancer patient: Case report and literature review. Infection 2003, 31, 421–424. [Google Scholar] [CrossRef]
  338. Fenner, B.J.; Kumar, A.; Tan, N.Y.Q.; Ang, M. Case of isolated Rhizobium radiobacter contact lens-related infectious keratitis: A plant microbe now emerging as a human pathogen. Am. J. Ophthalmol. Case Rep. 2019, 15, 100476. [Google Scholar] [CrossRef]
  339. Nuonming, P.; Khemthong, S.; Dokpikul, T.; Sukchawalit, R.; Mongkolsuk, S. Characterization and regulation of AcrABR, a RND-type multidrug efflux system, in Agrobacterium tumefaciens C58. Microbiol. Res. 2018, 214, 146–155. [Google Scholar] [CrossRef]
  340. Palumbo, J.D.; Kado, C.I.; Phillips, D.A. An isoflavonoid-inducible efflux pump in Agrobacterium tumefaciens is involved in competitive colonization of roots. J. Bacteriol. 1998, 180, 3107–3113. [Google Scholar] [CrossRef] [Green Version]
  341. Martinez, J.L.; Martinez-Suarez, J.; Culebras, E.; Perez-Diaz, J.C.; Baquero, F. Antibiotic inactivating enzymes from a clinical isolate of Agrobacterium radiobacter. J. Antimicrob. Chemother. 1989, 23, 283–284. [Google Scholar] [CrossRef]
  342. Lobaton, T.; Hoffman, I.; Vermeire, S.; Ferrante, M.; Verhaegen, J.; Van Assche, G. Aeromonas species: An opportunistic enteropathogen in patients with inflammatory bowel diseases? A single center cohort study. Inflamm. Bowel Dis. 2015, 21, 71–78. [Google Scholar] [CrossRef] [PubMed]
  343. Hofer, E.; Reis, C.M.; Theophilo, G.N.; Cavalcanti, V.O.; Lima, N.V.; Henriques Mde, F. Aeromonas associated with an acute diarrhea outbreak in Sao Bento do Una, Pernambuco. Rev. Soc. Bras. Med. Trop. 2006, 39, 217–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Hanninen, M.L.; Salmi, S.; Mattila, L.; Taipalinen, R.; Siitonen, A. Association of Aeromonas spp. with travellers′ diarrhoea in Finland. J. Med Microbiol. 1995, 42, 26–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Meng, C.Y.; Smith, B.L.; Bodhidatta, L.; Richard, S.A.; Vansith, K.; Thy, B.; Srijan, A.; Serichantalergs, O.; Mason, C.J. Etiology of diarrhea in young children and patterns of antibiotic resistance in Cambodia. Pediatric Infect. Dis. J. 2011, 30, 331–335. [Google Scholar] [CrossRef]
  346. Santos, Y.; Toranzo, A.E.; Barja, J.L.; Nieto, T.P.; Villa, T.G. Virulence properties and enterotoxin production of Aeromonas strains isolated from fish. Infect. Immun. 1988, 56, 3285–3293. [Google Scholar] [CrossRef] [Green Version]
  347. Soler, L.; Figueras, M.J.; Chacon, M.R.; Vila, J.; Marco, F.; Martinez-Murcia, A.J.; Guarro, J. Potential virulence and antimicrobial susceptibility of Aeromonas popoffii recovered from freshwater and seawater. FEMS Immunol. Med. Microbiol. 2002, 32, 243–247. [Google Scholar] [CrossRef]
  348. Kirov, S.M. The public health significance of Aeromonas spp. in foods. Int. J. Food Microbiol. 1993, 20, 179–198. [Google Scholar] [CrossRef]
  349. Nguyen, H.N.; Van, T.T.; Nguyen, H.T.; Smooker, P.M.; Shimeta, J.; Coloe, P.J. Molecular characterization of antibiotic resistance in Pseudomonas and Aeromonas isolates from catfish of the Mekong Delta, Vietnam. Vet. Microbiol. 2014, 171, 397–405. [Google Scholar] [CrossRef] [PubMed]
  350. Figueras, M.J.; Latif-Eugenin, F.; Ballester, F.; Pujol, I.; Tena, D.; Berg, K.; Hossain, M.J.; Beaz-Hidalgo, R.; Liles, M.R. ‘Aeromonas intestinalis’ and ‘Aeromonas enterica’ isolated from human faeces, ‘Aeromonas crassostreae’ from oyster and ‘Aeromonas aquatilis’ isolated from lake water represent novel species. New Microbes New Infect. 2017, 15, 74–76. [Google Scholar] [CrossRef]
  351. Bello-Lopez, J.M.; Cabrero-Martinez, O.A.; Ibanez-Cervantes, G.; Hernandez-Cortez, C.; Pelcastre-Rodriguez, L.I.; Gonzalez-Avila, L.U.; Castro-Escarpulli, G. Horizontal Gene Transfer and Its Association with Antibiotic Resistance in the Genus Aeromonas spp. Microorganisms 2019, 7, 363. [Google Scholar] [CrossRef] [Green Version]
  352. Yao, Z.; Sun, L.; Wang, Y.; Lin, L.; Guo, Z.; Li, D.; Lin, W.; Lin, X. Quantitative Proteomics Reveals Antibiotics Resistance Function of Outer Membrane Proteins in Aeromonas hydrophila. Front. Cell. Infect. Microbiol. 2018, 8, 390. [Google Scholar] [CrossRef] [Green Version]
  353. Joseph, S.W.; Daily, O.P.; Hunt, W.S.; Seidler, R.J.; Allen, D.A.; Colwell, R.R. Aeromonas primary wound infection of a diver in polluted waters. J. Clin. Microbiol. 1979, 10, 46–49. [Google Scholar] [CrossRef] [Green Version]
  354. Son, R.; Rusul, G.; Sahilah, A.M.; Zainuri, A.; Raha, A.R.; Salmah, I. Antibiotic resistance and plasmid profile of Aeromonas hydrophila isolates from cultured fish, Telapia (Telapia mossambica). Lett. Appl. Microbiol. 1997, 24, 479–482. [Google Scholar] [CrossRef] [Green Version]
  355. Figueira, V.; Vaz-Moreira, I.; Silva, M.; Manaia, C.M. Diversity and antibiotic resistance of Aeromonas spp. in drinking and waste water treatment plants. Water Res. 2011, 45, 5599–5611. [Google Scholar] [CrossRef] [PubMed]
  356. Morita, K.; Watanabe, N.; Kurata, S.; Kanamori, M. Beta-Lactam resistance of motile Aeromonas isolates from clinical and environmental sources. Antimicrob. Agents Chemother. 1994, 38, 353–355. [Google Scholar] [CrossRef] [Green Version]
  357. Starr, M.P.; Chatterjee, A.K. The genus Erwinia: Enterobacteria pathogenic to plants and animals. Annu. Rev. Microbiol. 1972, 26, 389–426. [Google Scholar] [CrossRef] [PubMed]
  358. Umenai, T.; Saitoh, Y.; Takano, S.; Shoji, E.; Tanaka, K.; Ishida, N. Significance of Erwinia in the vagina as causative agents of urinary tract infections. Tohoku J. Exp. Med. 1979, 129, 103–104. [Google Scholar] [CrossRef] [Green Version]
  359. Basset, A.; Khush, R.S.; Braun, A.; Gardan, L.; Boccard, F.; Hoffmann, J.A.; Lemaitre, B. The phytopathogenic bacteria Erwinia carotovora infects Drosophila and activates an immune response. Proc. Natl. Acad. Sci. USA 2000, 97, 3376–3381. [Google Scholar] [CrossRef]
  360. O’Hara, C.M.; Steigerwalt, A.G.; Hill, B.C.; Miller, J.M.; Brenner, D.J. First report of a human isolate of Erwinia persicinus. J. Clin. Microbiol. 1998, 36, 248–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  361. Gilardi, G.L.; Bottone, E.; Birnbaum, M. Unusual fermentative, gram-negative bacilli isolated from clinical specimens. I. Characterization of Erwinia strains of the “lathyri-herbicola group”. Appl. Microbiol. 1970, 20, 151–155. [Google Scholar] [CrossRef]
  362. Wechsler, A.; Bottone, E.; Lasser, R.; Korenman, G. Brain abscess caused by an Erwinia species. Report of a case and review of the literature. Am. J. Med. 1971, 51, 680–684. [Google Scholar] [CrossRef]
  363. Prod’homme, M.; Micol, L.A.; Weitsch, S.; Gassend, J.L.; Martinet, O.; Bellini, C. Cutaneous infection and bactaeremia caused by Erwinia billingiae: A case report. New Microbes New Infect. 2017, 19, 134–136. [Google Scholar] [CrossRef]
  364. Bonnet, I.; Bozzi, B.; Fourniols, E.; Mitrovic, S.; Soulier-Escrihuela, O.; Brossier, F.; Sougakoff, W.; Robert, J.; Jauréguiberry, S.; Aubry, A. Erwinia billingiae as Unusual Cause of Septic Arthritis, France, 2017. Emerg. Infect. Dis. 2019, 25, 1587–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  365. Sundin, G.W.; Wang, N. Antibiotic Resistance in Plant-Pathogenic Bacteria. Annu. Rev. Phytopathol. 2018, 56, 161–180. [Google Scholar] [CrossRef]
  366. Manulis, S.; Kleitman, F.; Shtienberg, D.; Shwartz, H.; Oppenheim, D.; Zilberstaine, M.; Shabi, E. Changes in the Sensitivity of Erwinia amylovora Populations to Streptomycin and Oxolinic Acid in Israel. Plant Dis. 2003, 87, 650–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  367. Nadarasah, G.; Stavrinides, J. Quantitative evaluation of the host-colonizing capabilities of the enteric bacterium Pantoea using plant and insect hosts. Microbiology (Reading) 2014, 160 Pt 3, 602–615. [Google Scholar] [CrossRef]
  368. Walterson, A.M.; Stavrinides, J. Pantoea: Insights into a highly versatile and diverse genus within the Enterobacteriaceae. FEMS Microbiol. Rev. 2015, 39, 968–984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  369. Soutar, C.D.; Stavrinides, J. Molecular validation of clinical Pantoea isolates identified by MALDI-TOF. PLoS ONE 2019, 14, e0224731. [Google Scholar] [CrossRef] [Green Version]
  370. Johnson, K.B.; Stockwell, V.O.; Sawyer, T.L.; Sugar, D. Assessment of Environmental Factors Influencing Growth and Spread of Pantoea agglomerans on and Among Blossoms of Pear and Apple. Phytopathology 2000, 90, 1285–1294. [Google Scholar] [CrossRef] [Green Version]
  371. Pileggi, M.; Pileggi, S.A.; Olchanheski, L.R.; da Silva, P.A.; Munoz Gonzalez, A.M.; Koskinen, W.C.; Barber, B.; Sadowsky, M.J. Isolation of mesotrione-degrading bacteria from aquatic environments in Brazil. Chemosphere 2012, 86, 1127–1132. [Google Scholar] [CrossRef] [Green Version]
  372. Guevarra, R.B.; Magez, S.; Peeters, E.; Chung, M.S.; Kim, K.H.; Radwanska, M. Comprehensive genomic analysis reveals virulence factors and antibiotic resistance genes in Pantoea agglomerans KM1, a potential opportunistic pathogen. PLoS ONE 2021, 16, e0239792. [Google Scholar] [CrossRef] [PubMed]
  373. Mehar, V.; Yadav, D.; Sanghvi, J.; Gupta, N.; Singh, K. Pantoea dispersa: An unusual cause of neonatal sepsis. Braz. J. Infect. Dis. Off. Publ. Braz. Soc. Infect. Dis. 2013, 17, 726–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  374. Hagiya, H.; Otsuka, F. Pantoea dispersa bacteremia caused by central line-associated bloodstream infection. Braz. J. Infect. Dis. Off. Publ. Braz. Soc. Infect. Dis. 2014, 18, 696–697. [Google Scholar] [CrossRef] [Green Version]
  375. Brady, C.L.; Cleenwerck, I.; Venter, S.N.; Engelbeen, K.; De Vos, P.; Coutinho, T.A. Emended description of the genus Pantoea, description of four species from human clinical samples, Pantoea septica sp. nov., Pantoea eucrina sp. nov., Pantoea brenneri sp. nov. and Pantoea conspicua sp. nov., and transfer of Pectobacterium cypripedii (Hori 1911) Brenner et al., 1973 emend. Hauben et al. 1998 to the genus as Pantoea cypripedii comb. nov. Int. J. Syst. Evol. Microbiol. 2010, 60 Pt 10, 2430–2440. [Google Scholar]
  376. Büyükcam, A.; Tuncer, Ö.; Gür, D.; Sancak, B.; Ceyhan, M.; Cengiz, A.B.; Kara, A. Clinical and microbiological characteristics of Pantoea agglomerans infection in children. J. Infect. Public Health 2018, 11, 304–309. [Google Scholar] [CrossRef] [PubMed]
  377. Martinez, J.L. General principles of antibiotic resistance in bacteria. Drug Discov. Today. Technol. 2014, 11, 33–39. [Google Scholar] [CrossRef]
  378. Martinez, J.L.; Fajardo, A.; Garmendia, L.; Hernandez, A.; Linares, J.F.; Martinez-Solano, L.; Sanchez, M.B. A global view of antibiotic resistance. FEMS Microbiol. Rev. 2009, 33, 44–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  379. Fajardo, A.; Linares, J.F.; Martinez, J.L. Towards an ecological approach to antibiotics and antibiotic resistance genes. Clin. Microbiol. Infect. 2009, 15 (Suppl. S1), 14–16. [Google Scholar] [CrossRef] [PubMed]
  380. Baquero, F.; Alvarez-Ortega, C.; Martinez, J.L. Ecology and evolution of antibiotic resistance. Environ. Microbiol. Rep. 2009, 1, 469–476. [Google Scholar] [CrossRef]
  381. Macinga, D.R.; Rather, P.N. The chromosomal 2′-N-acetyltransferase of Providencia stuartii: Physiological functions and genetic regulation. Front Biosci. 1999, 4, D132–D140. [Google Scholar] [CrossRef] [Green Version]
  382. Matz, C.; Deines, P.; Jurgens, K. Phenotypic variation in Pseudomonas sp. CM10 determines microcolony formation and survival under protozoan grazing. FEMS Microbiol. Ecol. 2002, 39, 57–65. [Google Scholar] [CrossRef] [PubMed]
  383. Bianco, N.; Neshat, S.; Poole, K. Conservation of the multidrug resistance efflux gene oprM in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 1997, 41, 853–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  384. Poole, K. Multidrug efflux pumps and antimicrobial resistance in Pseudomonas aeruginosa and related organisms. J. Mol. Microbiol. Biotechnol. 2001, 3, 255–264. [Google Scholar] [PubMed]
  385. Rosenberg, E.Y.; Bertenthal, D.; Nilles, M.L.; Bertrand, K.P.; Nikaido, H. Bile salts and fatty acids induce the expression of Escherichia coli AcrAB multidrug efflux pump through their interaction with Rob regulatory protein. Mol. Microbiol. 2003, 48, 1609–1619. [Google Scholar] [CrossRef] [PubMed]
  386. Maseda, H.; Sawada, I.; Saito, K.; Uchiyama, H.; Nakae, T.; Nomura, N. Enhancement of the mexAB-oprM efflux pump expression by a quorum-sensing autoinducer and its cancellation by a regulator, MexT, of the mexEF-oprN efflux pump operon in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2004, 48, 1320–1328. [Google Scholar] [CrossRef] [Green Version]
  387. Prouty, A.M.; Brodsky, I.E.; Falkow, S.; Gunn, J.S. Bile-salt-mediated induction of antimicrobial and bile resistance in Salmonella typhimurium. Microbiology (Reading) 2004, 150 Pt 4, 775–783. [Google Scholar] [CrossRef]
  388. Ravirala, R.S.; Barabote, R.D.; Wheeler, D.M.; Reverchon, S.; Tatum, O.; Malouf, J.; Liu, H.; Pritchard, L.; Hedley, P.E.; Birch, P.R.; et al. Efflux pump gene expression in Erwinia chrysanthemi is induced by exposure to phenolic acids. Mol. Plant Microbe Interact. 2007, 20, 313–320. [Google Scholar] [CrossRef] [Green Version]
  389. Sawada, I.; Maseda, H.; Nakae, T.; Uchiyama, H.; Nomura, N. A quorum-sensing autoinducer enhances the mexAB-oprM efflux-pump expression without the MexR-mediated regulation in Pseudomonas aeruginosa. Microbiol. Immunol. 2004, 48, 435–439. [Google Scholar] [CrossRef] [Green Version]
  390. Vargas, P.; Felipe, A.; Michan, C.; Gallegos, M.T. Induction of Pseudomonas syringae pv. tomato DC3000 MexAB-OprM multidrug efflux pump by flavonoids is mediated by the repressor PmeR. Mol. Plant Microbe Interact. 2011, 24, 1207–1219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  391. Lin, J.; Cagliero, C.; Guo, B.; Barton, Y.W.; Maurel, M.C.; Payot, S.; Zhang, Q. Bile salts modulate expression of the CmeABC multidrug efflux pump in Campylobacter jejuni. J. Bacteriol. 2005, 187, 7417–7424. [Google Scholar] [CrossRef] [Green Version]
  392. Pletzer, D.; Weingart, H. Characterization and regulation of the resistance-nodulation-cell division-type multidrug efflux pumps MdtABC and MdtUVW from the fire blight pathogen Erwinia amylovora. BMC Microbiol. 2014, 14, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  393. Laudy, A.E. Non-antibiotics, Efflux Pumps and Drug Resistance of Gram-negative Rods. Pol. J. Microbiol. 2018, 67, 129–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Papadopoulos, C.J.; Carson, C.F.; Chang, B.J.; Riley, T.V. Role of the MexAB-OprM efflux pump of Pseudomonas aeruginosa in tolerance to tea tree (Melaleuca alternifolia) oil and its monoterpene components terpinen-4-ol, 1,8-cineole, and alpha-terpineol. Appl. Environ. Microbiol. 2008, 74, 1932–1935. [Google Scholar] [CrossRef] [Green Version]
  395. Barabote, R.D.; Johnson, O.L.; Zetina, E.; San Francisco, S.K.; Fralick, J.A.; San Francisco, M.J. Erwinia chrysanthemi tolC is involved in resistance to antimicrobial plant chemicals and is essential for phytopathogenesis. J. Bacteriol. 2003, 185, 5772–5778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  396. Pusey, P.L.; Stockwell, V.O.; Reardon, C.L.; Smits, T.H.; Duffy, B. Antibiosis activity of Pantoea agglomerans biocontrol strain E325 against Erwinia amylovora on apple flower stigmas. Phytopathology 2011, 101, 1234–1241. [Google Scholar] [CrossRef] [Green Version]
  397. Lamarche, M.G.; Deziel, E. MexEF-OprN Efflux Pump Exports the Pseudomonas Quinolone Signal (PQS) Precursor HHQ (4-hydroxy-2-heptylquinoline). PLoS ONE 2011, 6, e24310. [Google Scholar] [CrossRef] [PubMed]
  398. Olivares, J.; Alvarez-Ortega, C.; Linares, J.F.; Rojo, F.; Kohler, T.; Martinez, J.L. Overproduction of the multidrug efflux pump MexEF-OprN does not impair Pseudomonas aeruginosa fitness in competition tests, but produces specific changes in bacterial regulatory networks. Environ. Microbiol. 2012, 14, 1968–1981. [Google Scholar] [CrossRef] [PubMed]
  399. Aendekerk, S.; Diggle, S.P.; Song, Z.; Hoiby, N.; Cornelis, P.; Williams, P.; Camara, M. The MexGHI-OpmD multidrug efflux pump controls growth, antibiotic susceptibility and virulence in Pseudomonas aeruginosa via 4-quinolone-dependent cell-to-cell communication. Microbiology (Reading) 2005, 151 Pt 4, 1113–1125. [Google Scholar] [CrossRef] [Green Version]
  400. Bever, J.D.; Westover, K.M.; Antonovics, J. Incorporating the soil community into plant pop-ulation dynamics: The utility of the feedback approach. J. Ecol. 1997, 85, 561–563. [Google Scholar] [CrossRef]
  401. Walker, T.S.; Bais, H.P.; Grotewold, E.; Vivanco, J.M. Root exudation and rhizosphere biology. Plant Physiol. 2003, 132, 44–51. [Google Scholar] [CrossRef] [Green Version]
  402. Bever, J.D.; Platt, T.G.; Morton, E.R. Microbial population and community dynamics on plant roots and their feedbacks on plant communities. Annu. Rev. Microbiol. 2012, 66, 265–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  403. Pitman, A.R.; Jackson, R.W.; Mansfield, J.W.; Kaitell, V.; Thwaites, R.; Arnold, D.L. Exposure to host resistance mechanisms drives evolution of bacterial virulence in plants. Curr. Biol. 2005, 15, 2230–2235. [Google Scholar] [CrossRef] [Green Version]
  404. Zhou, H.; Morgan, R.L.; Guttman, D.S.; Ma, W. Allelic variants of the Pseudomonas syringae type III effector HopZ1 are differentially recognized by plant resistance systems. Mol. Plant Microbe Interact. 2009, 22, 176–189. [Google Scholar] [CrossRef] [Green Version]
  405. LaPointe, G.; Nautiyal, C.S.; Chilton, W.S.; Farrand, S.K.; Dion, P. Spontaneous mutation conferring the ability to catabolize mannopine in Agrobacterium tumefaciens. J. Bacteriol. 1992, 174, 2631–2639. [Google Scholar] [CrossRef] [Green Version]
  406. Kang, H.; Gross, D.C. Characterization of a resistance-nodulation-cell division transporter system associated with the syr-syp genomic island of Pseudomonas syringae pv. syringae. Appl. Environ. Microbiol. 2005, 71, 5056–5065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  407. Burse, A.; Weingart, H.; Ullrich, M.S. The phytoalexin-inducible multidrug efflux pump AcrAB contributes to virulence in the fire blight pathogen, Erwinia amylovora. Mol. Plant Microbe Interact. 2004, 17, 43–54. [Google Scholar] [CrossRef] [Green Version]
  408. Takeshima, K.; Hidaka, T.; Wei, M.; Yokoyama, T.; Minamisawa, K.; Mitsui, H.; Itakura, M.; Kaneko, T.; Tabata, S.; Saeki, K.; et al. Involvement of a novel genistein-inducible multidrug efflux pump of Bradyrhizobium japonicum early in the interaction with Glycine max (L.) Merr. Microbes Environ. 2013, 28, 414–421. [Google Scholar] [CrossRef] [Green Version]
  409. Brown, D.G.; Swanson, J.K.; Allen, C. Two host-induced Ralstonia solanacearum genes, acrA and dinF, encode multidrug efflux pumps and contribute to bacterial wilt virulence. Appl. Environ. Microbiol. 2007, 73, 2777–2786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  410. Rossbach, S.; Kunze, K.; Albert, S.; Zehner, S.; Gottfert, M. The Sinorhizobium meliloti EmrAB efflux system is regulated by flavonoids through a TetR-like regulator (EmrR). Mol. Plant Microbe Interact. 2014, 27, 379–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  411. Poole, K. Multidrug resistance in Gram-negative bacteria. Curr. Opin. Microbiol. 2001, 4, 500–508. [Google Scholar] [CrossRef]
  412. Rodriguez-Martinez, J.M.; Poirel, L.; Nordmann, P. Molecular epidemiology and mechanisms of carbapenem resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2009, 53, 4783–4788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  413. Masuda, N.; Sakagawa, E.; Ohya, S.; Gotoh, N.; Tsujimoto, H.; Nishino, T. Substrate specificities of MexAB-OprM, MexCD-OprJ, and MexXY-oprM efflux pumps in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2000, 44, 3322–3327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  414. Hernando-Amado, S.; Blanco, P.; Alcalde-Rico, M.; Corona, F.; Reales-Calderon, J.A.; Sanchez, M.B.; Martinez, J.L. Multidrug efflux pumps as main players in intrinsic and acquired resistance to antimicrobials. Drug Resist. Updat. Rev. Comment. Antimicrob. Anticancer. Chemother. 2016, 28, 13–27. [Google Scholar] [CrossRef]
  415. Ziha-Zarifi, I.; Llanes, C.; Kohler, T.; Pechere, J.C.; Plesiat, P. In vivo emergence of multidrug-resistant mutants of Pseudomonas aeruginosa overexpressing the active efflux system MexA-MexB-OprM. Antimicrob. Agents Chemother. 1999, 43, 287–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  416. Vargas, P.; Farias, G.A.; Nogales, J.; Prada, H.; Carvajal, V.; Baron, M.; Rivilla, R.; Martin, M.; Olmedilla, A.; Gallegos, M.T. Plant flavonoids target Pseudomonas syringae pv. tomato DC3000 flagella and type III secretion system. Environ. Microbiol. Rep. 2013, 5, 841–850. [Google Scholar] [CrossRef]
  417. Chatterjee, A.; Cui, Y.; Yang, H.; Collmer, A.; Alfano, J.R.; Chatterjee, A.K. GacA, the response regulator of a two-component system, acts as a master regulator in Pseudomonas syringae pv. tomato DC3000 by controlling regulatory RNA, transcriptional activators, and alternate sigma factors. Mol. Plant Microbe Interact. 2003, 16, 1106–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  418. Al-Karablieh, N.; Weingart, H.; Ullrich, M.S. The outer membrane protein TolC is required for phytoalexin resistance and virulence of the fire blight pathogen Erwinia Amylovora. Microb. Biotechnol. 2009, 2, 465–475. [Google Scholar] [CrossRef] [PubMed]
  419. Chatnaparat, T.; Prathuangwong, S.; Lindow, S.E. Global Pattern of Gene Expression of Xanthomonas axonopodis pv. glycines Within Soybean Leaves. Mol. Plant Microbe Interact. 2016, 29, 508–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  420. Laborda, P.; Alcalde-Rico, M.; Chini, A.; Martínez, J.L.; Hernando-Amado, S. Discovery of inhibitors of Pseudomonas aeruginosa virulence through the search for natural-like compounds with a dual role as inducers and substrates of efflux pumps. Environ. Microbiol. 2021. [Google Scholar] [CrossRef]
  421. Rasmann, S.; Turlings, T.C. Root signals that mediate mutualistic interactions in the rhizosphere. Curr. Opin. Plant Biol. 2016, 32, 62–68. [Google Scholar] [CrossRef] [Green Version]
  422. Peters, N.K.; Frost, J.W.; Long, S.R. A plant flavone, luteolin, induces expression of Rhizobium meliloti nodulation genes. Science 1986, 233, 977–980. [Google Scholar] [CrossRef]
  423. Santos, M.R.; Marques, A.T.; Becker, J.D.; Moreira, L.M. The Sinorhizobium meliloti EmrR regulator is required for efficient colonization of Medicago sativa root nodules. Mol. Plant Microbe Interact. 2014, 27, 388–399. [Google Scholar] [CrossRef] [Green Version]
  424. Bais, H.P.; Weir, T.L.; Perry, L.G.; Gilroy, S.; Vivanco, J.M. The role of root exudates in rhizosphere interactions with plants and other organisms. Annu. Rev. Plant Biol. 2006, 57, 233–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  425. Neal, A.L.; Ahmad, S.; Gordon-Weeks, R.; Ton, J. Benzoxazinoids in root exudates of maize attract Pseudomonas putida to the rhizosphere. PLoS ONE 2012, 7, e35498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  426. De Kievit, T.R.; Parkins, M.D.; Gillis, R.J.; Srikumar, R.; Ceri, H.; Poole, K.; Iglewski, B.H.; Storey, D.G. Multidrug efflux pumps: Expression patterns and contribution to antibiotic resistance in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 2001, 45, 1761–1770. [Google Scholar] [CrossRef] [Green Version]
  427. Evans, K.; Passador, L.; Srikumar, R.; Tsang, E.; Nezezon, J.; Poole, K. Influence of the MexAB-OprM multidrug efflux system on quorum sensing in Pseudomonas aeruginosa. J. Bacteriol. 1998, 180, 5443–5447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  428. Pearson, J.P.; Van Delden, C.; Iglewski, B.H. Active efflux and diffusion are involved in transport of Pseudomonas aeruginosa cell-to-cell signals. J. Bacteriol. 1999, 181, 1203–1210. [Google Scholar] [CrossRef] [Green Version]
  429. Minagawa, S.; Inami, H.; Kato, T.; Sawada, S.; Yasuki, T.; Miyairi, S.; Horikawa, M.; Okuda, J.; Gotoh, N. RND type efflux pump system MexAB-OprM of Pseudomonas aeruginosa selects bacterial languages, 3-oxo-acyl-homoserine lactones, for cell-to-cell communication. BMC Microbiol. 2012, 12, 70. [Google Scholar] [CrossRef] [Green Version]
  430. Alcalde-Rico, M.; Olivares-Pacheco, J.; Alvarez-Ortega, C.; Camara, M.; Martinez, J.L. Role of the Multidrug Resistance Efflux Pump MexCD-OprJ in the Pseudomonas aeruginosa Quorum Sensing Response. Front. Microbiol. 2018, 9, 2752. [Google Scholar] [CrossRef]
  431. Alcalde-Rico, M.; Olivares-Pacheco, J.; Halliday, N.; Camara, M.; Martinez, J.L. The impaired quorum sensing response of Pseudomonas aeruginosa MexAB-OprM efflux pump overexpressing mutants is not due to non-physiological efflux of 3-oxo-C12-HSL. Environ. Microbiol. 2020, 22, 5167–5188. [Google Scholar] [CrossRef] [PubMed]
  432. Fray, R.G. Altering plant-microbe interaction through artificially manipulating bacterial quorum sensing. Ann. Bot. 2002, 89, 245–253. [Google Scholar] [CrossRef] [Green Version]
  433. Bauer, W.D.; Mathesius, U. Plant responses to bacterial quorum sensing signals. Curr. Opin. Plant Biol. 2004, 7, 429–433. [Google Scholar] [CrossRef]
  434. Teplitski, M.; Robinson, J.B.; Bauer, W.D. Plants secrete substances that mimic bacterial N-acyl homoserine lactone signal activities and affect population density-dependent behaviors in associated bacteria. Mol. Plant Microbe Interact. 2000, 13, 637–648. [Google Scholar] [CrossRef] [Green Version]
  435. Koh, C.L.; Sam, C.K.; Yin, W.F.; Tan, L.Y.; Krishnan, T.; Chong, Y.M.; Chan, K.G. Plant-derived natural products as sources of anti-quorum sensing compounds. Sensors 2013, 13, 6217–6228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  436. Manefield, M.; de Nys, R.; Naresh, K.; Roger, R.; Givskov, M.; Peter, S.; Kjelleberg, S. Evidence that halogenated furanones from Delisea pulchra inhibit acylated homoserine lactone (AHL)-mediated gene expression by displacing the AHL signal from its receptor protein. Microbiology 1999, 145 Pt 2, 283–291. [Google Scholar] [CrossRef] [Green Version]
  437. Uroz, S.; Dessaux, Y.; Oger, P. Quorum sensing and quorum quenching: The yin and yang of bacterial communication. Chembiochem 2009, 10, 205–216. [Google Scholar] [CrossRef] [PubMed]
  438. Dong, Y.H.; Wang, L.H.; Xu, J.L.; Zhang, H.B.; Zhang, X.F.; Zhang, L.H. Quenching quorum-sensing-dependent bacterial infection by an N-acyl homoserine lactonase. Nature 2001, 411, 813–817. [Google Scholar] [CrossRef] [PubMed]
  439. Burse, A.; Weingart, H.; Ullrich, M.S. NorM, an Erwinia amylovora multidrug efflux pump involved in in vitro competition with other epiphytic bacteria. Appl. Environ. Microbiol. 2004, 70, 693–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  440. Piddock, L.J. Multidrug-resistance efflux pumps—Not just for resistance. Nat. Rev. Microbiol. 2006, 4, 629–636. [Google Scholar] [CrossRef] [PubMed]
  441. Kim, I.Y.; Pusey, P.L.; Zhao, Y.; Korban, S.S.; Choi, H.; Kim, K.K. Controlled release of Pantoea agglomerans E325 for biocontrol of fire blight disease of apple. J. Control. Release Off. J. Control. Release Soc. 2012, 161, 109–115. [Google Scholar] [CrossRef] [PubMed]
  442. Jacob, F. Evolution and tinkering. Science 1977, 196, 1161–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  443. Kohler, T.; Michea-Hamzehpour, M.; Henze, U.; Gotoh, N.; Curty, L.K.; Pechere, J.C. Characterization of MexE-MexF-OprN, a positively regulated multidrug efflux system of Pseudomonas aeruginosa. Mol. Microbiol. 1997, 23, 345–354. [Google Scholar] [CrossRef] [PubMed]
  444. Wu, C.J.; Huang, Y.W.; Lin, Y.T.; Ning, H.C.; Yang, T.C. Inactivation of SmeSyRy Two-Component Regulatory System Inversely Regulates the Expression of SmeYZ and SmeDEF Efflux Pumps in Stenotrophomonas maltophilia. PLoS ONE 2016, 11, e0160943. [Google Scholar] [CrossRef] [PubMed]
  445. Alonso, A.; Martinez, J.L. Expression of multidrug efflux pump SmeDEF by clinical isolates of Stenotrophomonas maltophilia. Antimicrob. Agents Chemother. 2001, 45, 1879–1881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  446. Blanco, P.; Corona, F.; Martinez, J.L. Mechanisms and phenotypic consequences of acquisition of tigecycline resistance by Stenotrophomonas maltophilia. J. Antimicrob. Chemother. 2019, 74, 3221–3230. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the main elements involved in intrinsic and acquired antibiotic resistance in Pseudomonas aeruginosa. P. aeruginosa possesses a remarkable intrinsic resistance to antibiotics caused, among other factors, by the production of antibiotic-modifying enzymes (e.g., β-lactamase AmpC), low outer membrane permeability and a great amount of multidrug resistance (MDR) efflux pumps like MexAB-OprM. Antibiotic resistance level may increase by chromosomal mutations in genes encoding negative regulators of the above-described intrinsic resistance determinants, such as genetic modifications within mexR or ampR, which boost the expression of mexAB-oprM and ampC, respectively. The modification of the antibiotic target is also a frequent mechanism for acquiring antibiotic resistance in P. aeruginosa, as the increased resistance to quinolones by mutations in gyrases encoded by gyrA or gyrB. Alternatively, this bacterium is also able to acquire novel ARGs, which are located in mobile elements, such as plasmids or integrative and conjugative elements (ICEs). It has been stated that bacteriophages might also be involved in the acquisition of ARGs, but the role of these genetic elements in the spread of resistance in P. aeruginosa is not yet fully understood.
Figure 1. Schematic representation of the main elements involved in intrinsic and acquired antibiotic resistance in Pseudomonas aeruginosa. P. aeruginosa possesses a remarkable intrinsic resistance to antibiotics caused, among other factors, by the production of antibiotic-modifying enzymes (e.g., β-lactamase AmpC), low outer membrane permeability and a great amount of multidrug resistance (MDR) efflux pumps like MexAB-OprM. Antibiotic resistance level may increase by chromosomal mutations in genes encoding negative regulators of the above-described intrinsic resistance determinants, such as genetic modifications within mexR or ampR, which boost the expression of mexAB-oprM and ampC, respectively. The modification of the antibiotic target is also a frequent mechanism for acquiring antibiotic resistance in P. aeruginosa, as the increased resistance to quinolones by mutations in gyrases encoded by gyrA or gyrB. Alternatively, this bacterium is also able to acquire novel ARGs, which are located in mobile elements, such as plasmids or integrative and conjugative elements (ICEs). It has been stated that bacteriophages might also be involved in the acquisition of ARGs, but the role of these genetic elements in the spread of resistance in P. aeruginosa is not yet fully understood.
Ijms 22 08080 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sanz-García, F.; Gil-Gil, T.; Laborda, P.; Ochoa-Sánchez, L.E.; Martínez, J.L.; Hernando-Amado, S. Coming from the Wild: Multidrug Resistant Opportunistic Pathogens Presenting a Primary, Not Human-Linked, Environmental Habitat. Int. J. Mol. Sci. 2021, 22, 8080. https://doi.org/10.3390/ijms22158080

AMA Style

Sanz-García F, Gil-Gil T, Laborda P, Ochoa-Sánchez LE, Martínez JL, Hernando-Amado S. Coming from the Wild: Multidrug Resistant Opportunistic Pathogens Presenting a Primary, Not Human-Linked, Environmental Habitat. International Journal of Molecular Sciences. 2021; 22(15):8080. https://doi.org/10.3390/ijms22158080

Chicago/Turabian Style

Sanz-García, Fernando, Teresa Gil-Gil, Pablo Laborda, Luz E. Ochoa-Sánchez, José L. Martínez, and Sara Hernando-Amado. 2021. "Coming from the Wild: Multidrug Resistant Opportunistic Pathogens Presenting a Primary, Not Human-Linked, Environmental Habitat" International Journal of Molecular Sciences 22, no. 15: 8080. https://doi.org/10.3390/ijms22158080

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop