Next Article in Journal
Decoding the Microbiome’s Influence on Rheumatoid Arthritis
Next Article in Special Issue
Chronic Granulomatous Disease (CGD): Commonly Associated Pathogens, Diagnosis and Treatment
Previous Article in Journal
Comprehensive Response of Rhodosporidium kratochvilovae to Glucose Starvation: A Transcriptomics-Based Analysis
Previous Article in Special Issue
International Clones of High Risk of Acinetobacter Baumannii—Definitions, History, Properties and Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment

1
Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland
2
Department of Analytical, Biopharmaceutical and Medical Sciences, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(9), 2169; https://doi.org/10.3390/microorganisms11092169
Submission received: 28 July 2023 / Revised: 23 August 2023 / Accepted: 25 August 2023 / Published: 28 August 2023
(This article belongs to the Special Issue Latest Review Papers in Medical Microbiology 2023)

Abstract

:
Urinary tract infections (UTIs) are among the most common bacterial infections, especially among women and older adults, leading to a significant global healthcare cost burden. Uropathogenic Escherichia coli (UPEC) are the most common cause and accounts for the majority of community-acquired UTIs. Infection by UPEC can cause discomfort, polyuria, and fever. More serious clinical consequences can result in urosepsis, kidney damage, and death. UPEC is a highly adaptive pathogen which presents significant treatment challenges rooted in a complex interplay of molecular factors that allow UPEC to evade host defences, persist within the urinary tract, and resist antibiotic therapy. This review discusses these factors, which include the key genes responsible for adhesion, toxin production, and iron acquisition. Additionally, it addresses antibiotic resistance mechanisms, including chromosomal gene mutations, antibiotic deactivating enzymes, drug efflux, and the role of mobile genetic elements in their dissemination. Furthermore, we provide a forward-looking analysis of emerging alternative therapies, such as phage therapy, nano-formulations, and interventions based on nanomaterials, as well as vaccines and strategies for immunomodulation. This review underscores the continued need for research into the molecular basis of pathogenesis and antimicrobial resistance in the treatment of UPEC, as well as the need for clinically guided treatment of UTIs, particularly in light of the rapid spread of multidrug resistance.

1. Introduction

Urinary tract infections are one of the most common bacterial infections, with an estimated 400 million cases and 230,000 deaths worldwide in 2019 [1]. Some 50% of women will experience a UTI at least once in their lifetime, with recurrence being common [2]. Furthermore, UTIs are one of the most common infections reported in older adults, being second only to respiratory infections in hospitalised patients over 65 [3]. The economic burden of UTIs is high, representing 6% of medical visits in the United States and costing approximately USD 1.6 billion annually [4]. A European study which sought to determine the cost associated with complicated urinary tract infection in eight countries with a high prevalence of multidrug-resistant Gram-negative bacteria found that the average cost of treatment was EUR 5700 per case [5]. An Irish study from 2014 estimated the primary care cost of UTI in Ireland to be EUR 19.2 million annually [6]. UTI infections fall into distinct clinical categories as defined by the European Association of Urology (EAU), which in turn guides treatment [7]. Uncomplicated UTIs are described as infections involving non-pregnant, pre-menopausal women with no known relevant urological abnormalities or comorbidities, while complicated UTIs are those affecting men, pregnant women, patients with anatomical or functional abnormalities, renal disease, indwelling urinary catheters and those with certain immunosuppressive co-morbidities [7]. A recurrent UTI (R-UTI) is defined as two or more uncomplicated or complicated UTI infections in six months, or three infections within one year that are new infections with separate organisms [8]. Certain conditions or abnormalities may predispose a person to recurrent infection. This can occur due to relapse and infection by the same persistent organism following unsuccessful treatment, but in the majority of cases it results from infection with a separate organism, post the resolution of symptoms [9].
The main cause of UTIs is infection by uropathogenic Escherichia coli (UPEC), responsible for 75% of uncomplicated UTIs and 65% of complicated UTIs [10]. UPEC form part of a group of pathogenic E. coli known as extraintestinal pathogenic E. coli (ExPEC), a collection of four pathotypes that are each classified by their respective site of isolation, and have adopted virulence mechanisms that allow them to proliferate and cause disease in host sites outside of the intestinal tract [11,12]. ExPEC are separable from diarrheagenic E. coli (DEC), a group of six E. coli pathotypes which cause intestinal infections, and each possess a different combination of virulence characteristics [13]. Infection begins when bacteria that reside in the gut enter the urethra and colonise epithelial cells and eventually migrate upwards to colonise the bladder. The initiation of the host’s innate immune response to infection represents a complex cascade of events. Central to this response are pattern recognition receptors (PRRs). Notably among this family are Toll-like receptors (TLRs), which recognise pathogen-associated molecular patterns (PAMPs) [14]. TLRs are expressed across a number of cell types, including immune cells such as NK cells, macrophages, and leukocytes, as well as non-immune cells such as bladder, ureter, and kidney urothelial cells [15,16]. The 10 TLRs (TRL1-10) which are found in humans are distinguishable by the ligands they recognise and the resulting signalling pathways they activate [17]. Critical to the host’s immune response during invasion by UPEC is TLR4, the sensing receptor for Gram-negative lipopolysaccharide (LPS) [18]. Upon detection of LPS, TLR-4 activates the intracellular signalling pathway NF-κB which regulates the expression of many proinflammatory cytokines and chemokines as well as leukocyte attracting chemoattractants [19,20]. Further inflammation is stimulated by TLR-5, which responds to the presence of bacteria flagellin [21]. While the locally attracted phagocytes are effective at clearing extracellular pathogens, TLR-4 additionally mediates the expulsion of intracellular pathogens through cAMP-dependent exocytosis from bladder epithelial cells [22].
Nevertheless, UPEC have acquired an array of virulence factors to circumvent host immune mechanisms and expedite infection and disease. These factors include proteins related to motility and adhesion which allow bacterial cells to attach to host receptors [23,24]. Some of these adhesive surface proteins bind preferentially to kidney cells which predispose some UPEC to pyelonephritis [25]. Adhesions allow bacteria to resist expulsion via urination. As well as this, adherence to the uroepithelium is the first step in the formation of intracellular bacterial communities (IBCs), where bacterial cells can evade expulsion by TLR-4, proliferate within the uroepithelium, and infect nearby cells, leading to persistence within the urinary tract [26,27,28]. UPEC which more readily form biofilms are also more persistent and difficult to treat due to the innate ability of the biofilm to shield cells from antibiotic therapy [29]. Initial attachment to host cells through surface adhesions is an important determinant of a particular isolate’s ability to form a biofilm, and several studies have highlighted the differences in fimbria gene distribution between strong and weak biofilm formers [30,31]. Additionally, virulence factors and antibiotic resistance genes are more readily exchanged among bacteria while in a biofilm [32]. While adhesions play a significant role as fitness factors for UPEC during urinary tract colonisation, similarly, iron-chelating agents such as siderophores are crucial. These secreted molecules enable UPEC to sequester iron from the urinary tract, a vital element for various bacterial cell processes that is scarce in this specific environmental niche [33]. Genes which encode toxins are considered important virulence factors not only because they induce damage and morphological changes in host cells, but because they also assist in the evasion of the host’s immune defences [34,35,36]. The E. coli genome is highly plastic, and virulence factors can be transferred between strains by horizontal gene transfer, on large pathogenicity islands (PAI’s) within extrachromosomal plasmids [37,38] The transfer of virulence factors between pathotypes can result in hybrid strains, which include those that, although isolated from urinary tract infections and possess virulence factors associated with UPEC, also possess genes which are definitive of DEC pathotypes [39,40,41]. For example, UPEC/DEC strains have been isolated from urinary tract infections which carry the aggR gene, while others which were found to carry the eae gene have been reported [40,41]. The eae gene mediates the attachment of enteropathogenic Escherichia coli (EPEC) to epithelial cells [42], while the aggR gene is a transcriptional regulator which controls the expression of multiple virulence factors associated with enteroaggregative Escherichia coli (EAEC) [43]. These hybrids may have the ability to cause disease at multiple sites of infection. Some studies have looked to determine the frequency by which hybrid UPEC are isolated from UTIs, with reported figures between 2% and 10.5% [40,41,44].
When studying the genetics of E. coli, they can be divided using multiplex PCR into 8 major phylogroups, those being A, B1, B2, C, D, E, F, and Escherichia cryptic clade I [45]. The distribution of these groups varies between locations, between clinical and commensal strains, animal or human carriage, and distribution of resistance genes. A meta-analysis of human E. coli isolates from 26 different countries found that phylogroup A was the most prominent, with a median result of 36%, followed by B2 at 20%, and B1 at 16.4%. [46]. Virulent extra-intestinal strains studied from samples taken from south India show that the groups B2 and D are most common [47]. Specifically for UPEC, a meta-analysis of samples from Iran revealed that the phylogroup B2 is most prominent at 32%, while D, B1 and A make up 26%, 18%, and 8%, respectively [48]. In addition to phylogenetic typing, antigen serotyping, specifically the O (lipopolysaccharide), H (flagellum), and K (capsule) antigens are also used frequently for characterisation [49]. The O antigen is particularly variable, and currently there are 186 recognized O serotypes of E. coli [49]. Among these, the majority of UPEC belong to serogroups O1, O2, O4, O6, O7, O8, O14, O15, O16, O18, O21, O22, O25, O75, and O83 [50,51,52,53,54]. Among these O25, O15, and O8 are more frequently detected, although specific distribution is subject to geographical location and patient population [52,53,54,55,56]. Apropos to this, numerous investigations have established associations linking virulence, antibiotic resistance, and distinct serogroups [52,53,54,55,56], underscoring the utility of this serotyping approach for epidemiological surveillance across diverse locales.
For the treatment of uncomplicated UTIs, the EAU recommends the use of fosfomycin trometamol, nitrofurantoin, and pivmecillinam, while cephalosporins are recommended as alternative therapy (Table 1). Trimethoprim/trimethoprim-sulfamethoxazole is recommended if local resistance rates are <20% and it is also recommended for the treatment of men, with fluoroquinolones suggested in accordance with local susceptibility testing. For complicated UTIs it is recommended to use amoxicillin plus an aminoglycoside or a second-generation cephalosporin plus an aminoglycoside. Intravenous third-generation cephalosporins are advised for the empirical treatment of symptomatic complicated UTIs (Table 1). It is also recommended that ciprofloxacin and other fluoroquinolones not be used in patients from urology departments or in cases where the patient has used fluroquinolones in the last six months [7]. It has been shown that laboratory guided treatment is preferable to empirical treatment, as national or regional guidelines may not accurately reflect actual resistance rates in specific geographical locations [57].

2. Virulence Factors

The ability of UPEC to colonise the bladder and resist antibiotic therapy is attributed to several virulence factors, many of which are also related to biofilm formation [26,58,59]. These include adhesion factors such as Type 1 fimbriae, P fimbriae, curli fibres, S fimbriae, F1C fimbriae, Dr fimbriae, afimbrial adhesins, and PapC [60,61]. These adhesion factors enable UPEC to adhere to and invade host cells and tissues, evade the host immune response, and colonise the urinary tract [62,63]. In addition, the extracellular matrix of UPEC biofilms is composed mainly of amyloid curli fibres, which are encoded by the curli operon [64]. Curli fibres play a crucial role in UPEC biofilm formation and the extracellular matrix they compose provides protection from environmental stresses such as antibiotics, host defences, and nutrient deprivation [65]. Toxins such as α-hemolysin, cytotoxic necrotizing factor, and serine protease autotransporter can cause tissue damage, proteolytically cleave immune cell complexes, and trigger morphological changes in host cells which facilitate bacterial persistence [34,35,36].
Moreover, the ability of UPEC to acquire iron is highly dependent on siderophore-iron transporter proteins [33]. These proteins enable UPEC to scavenge iron from the host and other sources, which is essential for bacterial growth and survival in the environment of the urinary tract where iron levels are infinitesimal [33]. A representative set of the virulence factors that can be expressed by UPEC are shown in Figure 1.
Virulence factors such as adhesions and siderophores can coalesce into pathogenicity islands, be mobilised in transposons, and be transferred on plasmids [66]. This transfer of virulence factors enables UPEC to adapt and evolve rapidly in response to environmental pressures and facilitates the spread of antibiotic resistance genes among interspecies bacterial populations.

2.1. Adhesions

2.1.1. Type 1 Fimbriae

Fimbria adhesions are extracellular protein structures that allow UPEC to attach to bladder cells and resist expulsion from the urinary system through the flow of urine. The best defined and most common fimbria adhesions are type 1fimbria (T1F). Genomic analysis of UPEC populations have reported prevalence of T1F between 86–100% [60,67,68]. Apart from initial attachment, T1F have been shown to mediate the invasion of bladder and uroepithelial cells, forming IBCs which can form persistent reservoirs of infection, are more resistant to antibiotics, and are speculated to contribute to recurrent infections [69,70].
T1F are encoded by the fim operon found in the majority of UPEC, but importantly, is not always expressed [71]. This operon contains nine genes, fimA to fimI with fimA expressing the main structural element, and fimC and fimD playing roles in the transport and assembly of the fimbriae [72]. fimH is responsible for facilitating binding of the fimbriae to host receptors, and has specificity for D-mannose-containing structures on host cells, often uroplakins [23]. The D-mannose-specific structures required for attachment are more numerous in the lower urinary tract than in the kidneys, so it was long believed that T1F do not play a role in pyelonephritis; however, more recent data suggest that T1F are still expressed in kidney infections, though they play the role of facilitating inter-bacterial adhesion and biofilm formation, rather than attachment and host cell invasion [73].
Expression of T1F is controlled by a reversible process called phase variation; a process by which the expression of bacterial surface factors, primarily adhesions, can be switched on or off [74]. Phase variation, or antigenic variation, was first described as it related to T1F and can be considered a method of host immune evasion, allowing bacteria to lower its expression of antigenic surface adhesions when switched off as well as to prevent coexpression and interference between surface components [75,76]. Here, the process is controlled by an invertible DNA element containing the promoter sequence for fimA. This sequence, which contains two inverted repeats flanking an intervening DNA segment, lines up with fimA to allow site specific recombination mediated by a recombinase enzyme and promotes transcription [77]. Expression through phase variation is thought to be regulated through a complex network of systems which responds to stress, and are likely to become activated during infection because of the hostile environment of the host urinary tract exerting pressure on invading UPEC [78]. The gene regulation which occurs during this response is regulated by a system of small-nucleotide alarmones, pGpp, ppGpp, and pppGpp (guanosine tetra- and pentaphosphates) and is produced by the enzymes RelA and SpoT [79,80]. Higher intrinsic levels of (p)ppGpp in E. coli result in high alarmone concentration, and this system activates the regulator FimB that controls the phase variation of type 1 fimbriae in E. coli [81].

2.1.2. P Fimbriae

Like T1F, P fimbriae are extracellular protein appendages which play a role in the adherence of UPEC to uroepithelial cells [25]. They differ from type 1 fimbriae in that their adherence is mannose resistant, binding to globoseries glycosphingolipids as opposed to the mannose dependent adherence of type-1 fimbriae to mono-mannose structures [24]. They are also more associated with upper urinary tract infections and pyelonephritis (P fimbriae for pyelonephritis fimbriae) and are considered a particularly important virulence factor in UPEC, as they mediate attachment to epithelial cells in the kidneys and renal tubules [73,82]. P fimbriae are encoded chromosomally by the pap operon. The main fimbriae structure is encoded by papA, papEF encode adapter subunits and papG encodes the terminal adhesive protein [25]. Because of this, the papG gene is usually used as a molecular marker to determine the prevalence of P fimbriae among UPEC. Studies have reported the prevalence of the pap operon to be 14–28% in UPEC causing cystitis, 5–7% in commensal E. coli and 71–91% of UPEC isolated from patients with pyelonephritis [82,83]. Importantly, papG has four molecular variants, with distinct binding specificities. It follows therefore, that some of the molecular variants of papG create more virulent strains of UPEC for certain hosts, and several studies have sought to characterise the differences between these variants and the diseases they are associated with. For example, PapGII has been encountered more than other alleles in isolates from women with pyelonephritis, while PapGIII is found more commonly in UPEC which is causative of cystitis [84,85]. PapGI and PapGIV are encountered rarely and are less understood than the other two molecular confirmations. UPEC which are positive for the papGII gene has also been shown to be positive for additional virulence factors, which may contribute to its association with more severe, upper UTI [84].
Regulation of the pap operon by phase variation involves the formation of differential methylation patterns resulting from the competition between DNA adenine methylase (Dam) and leucine-responsive regulatory protein (Lrp) for binding to two sets of overlapping binding sites within the pap regulatory region [86]. The regulatory region is composed of two genes, the first is papI which activates the on-phase by interacting with Lrp-pap DNA complexes when a specific GATC sequences is methylated by Dam [87]. The second is papB which both activates papI transcription but also represses transcription when protein concentrations are high [88].

2.1.3. S Fimbriae

S fimbriae are mannose-resistant adhesions which are encoded by the sfa operon. This operon consists of nine genes. The SfaA protein forms the dominant sub-unit, while SfaG, SfaH, and SfaS form minor sub-units, and SfaF forms the outer membrane translocation assembly protein [89,90]. Regulatory proteins SfaB and SfaC control expression, and like other fimbriae are manged by phase variation, that in this case, is influenced by temperature, osmolarity, glucose concentration, and other environmental factors [91]. S fimbriae has specificity for alpha-sialyl-2,3-alpha-galactose residues present on the glycoproteins of urothelial tissues in the bladder and kidneys [89,92]. Apart from urinary tract infections, S fimbriae-positive E. coli have been known to cause meningitis in new-borns, likely due to their binding potential for sialo-glycoproteins which are also found on brain micro vascular endothelial cells [93]. Studies which used molecular analysis to determine the prevalence of the sfa operon in E. coli have reported figures between 42–87% [60,67,68,94]. The sfa operon has been found more commonly in strains which form strong biofilms in vitro, as determined by tissue culture plate methods [94,95].

2.1.4. The Afa/Dr Family of Fimbrial Surface Adhesins

The Afa/Dr family of adhesions are particularly associated with UPEC that cause cystitis in children, as well as pyelonephritis and recurrent urinary tract infections in young and pregnant women [96,97,98]. It is also regularly found in the genome of ST131, a particularly virulent UPEC clone [99]. Dr fimbria (diffuse adherence fimbria) are part of the Afa/Dr family of fimbrial surface adhesions and are separable from other adhesions by the wide range of binding specificity they possess. They can bind, firstly to the Dr blood group antigen on decay-accelerating factor (DAF), which is a glycoprotein present on the surface of uroepithelial cells [100]. Secondly, Dr fimbria can recognise carcinoembryonic antigen (CEA)-related cell adhesion molecules (CEACAMs) CEA, CEACAM1, CEACAM3, and CEACAM6 as binding receptors [101]. Interactions between Dr fimbria and these receptors have been shown to mediate internalisation and invasion of UPEC into the host epithelial cells [101,102].
The genes encoding afa/dr adhesions are composed of a primary transcriptional unit containing afaA which is a regulatory gene, afaB which encodes a chaperone protein, afaC which encodes an usher protein, afaE which encodes a major adhesion subunit, and a tip capping subunit protein encoded by afaD. This gene cluster is accompanied by a separate minor transcriptional gene; afaF [96]. Phase mediated expression of the afa/dr family adhesions is, like P fimbriae, mediated by differential methylation patterns caused by Dam and Lrp interactions in the afa/dr promoter region [103]. Activation, however, is carried out by integration host factor (IHF) and repression is mediated by H-NS, a histone-like nucleoid structuring protein [99]. Previously it has been shown that insertion sequences (IS) elements when inserted into the regulatory region of the Afa/dr operon cause increased expression in a ST131 UPEC clone [99].

2.1.5. Non-Fimbrial Adhesins

Repeat-in-toxins (RTX) are a family of proteins usually transported to the cell surface through the type I secretory system [104]. They include toxins such as cytolysins and hemolysin, but also non-fimbrial adhesions [105]. Type one secretion A (tosA) is one such non fimbrial adhesion, encoded by the tosRCBDAEF operon [106]. Expressed on the cell surface, this adhesion can bind to human kidney and epithelial cells in the upper urinary tract [106]. The operon is regulated by TosR, with assistance from the global regulatory proteins H-NS and Lrp and its expression involves cross talk with the regulatory systems of other fimbrial adhesions, including Pap and Foc, allowing for cell surface organisation of both fimbrial and non-fimbrial adhesions [107]. Over expression of TosA has been shown to increase biofilm formation in vitro, in both LB broth and in human urine [107].

2.2. Curli

Curli is a functional amyloid protein that is an essential component of the biofilm matrix in many bacterial species, including E. coli [108]. Curli fibres are formed by the self-assembly of the major curli subunit protein, CsgA, which polymerises into fibrils on the bacterial cell surface upon contact with the minor subunit protein CsgB [109]. Curli production is regulated by the transcription factor CsgD, which activates the expression of the curli biosynthesis genes, which include csgA, csgB, and csgG, as well as genes involved in nucleotide biosynthesis and other metabolic pathways [65]. CsgE, CsgF, and CsgG proteins comprise the outer membrane curli secretion system, with CsgG forming a pore like outer membrane structure and CsgE and CsgF functioning as chaperones for secreted curli fibers [110].
The curli biosynthesis pathway in E. coli is particularly sensitive to changes in nucleotide biosynthesis, as disruption of the de novo purine biosynthetic pathway leads to a reduction in the intracellular level of cyclic-di-GMP, a secondary messenger molecule that is required for the activation of the csgD gene [111]. Cyclic-di-GMP is synthesised by diguanylate cyclases and degraded by phosphodiesterases, and its intracellular level is tightly regulated by a network of enzymes and signalling pathways [112]. The disruption of purine biosynthesis leads to a decrease in the intracellular level of cyclic-di-GMP, which in turn leads to a reduction in curli production and biofilm formation [64]. A study which created transposon mutants found that a strain with an insertion in the purL gene, resulted in a knockout variant which had reduced curli production and diminished biofilm formation [113]. The purL gene codes for formyl-glycinamide ribonucleotide amido-transferase (FGAR-AT), which is an enzyme that catalyses the fourth step of the purine biosynthetic pathway [114].
The involvement of the CsgD regulator in extracellular matrix production induced by extracellular nitrate suggests that the regulation of biofilm formation is also linked to the availability of nutrients in the bacterial environment. This was demonstrated by the creation of mutant strains lacking the membrane bound nitrate reductases NarGHJI and NarZYWV and produced increased biofilm as a result [115].

2.3. Biofilm Formation

Biofilm formation is a complex process that involves various genes and regulatory mechanisms. The fimbrial genes fimB, fimE, and the curli assembly component csgE are crucial for biofilm formation at all stages. However, during the initial 12 h of formation, genes related to gene transcription and motility play a more significant role along with these adhesion genes [116]. Several studies have investigated the relationship between phylogenetic groups and biofilm formation. Phylogroups B2 and D are most frequently associated with stronger biofilm formation or exhibit multidrug resistance and carry a high percentage of fimbrial, iron uptake, and toxin genes [117,118]. Moreover, one study reported no correlation between phylogenetic classification and biofilm formation, but a correlation was observed between antibiotic resistance and strong biofilm formation irrespective of phylogroup [81]. More generally, the biofilm formation of UPEC is variable between clonally distinct strains, and can include both weak and strong formers [119].
A study which characterised multidrug-resistant UPEC by phylogenic group found that adhesion genes were more commonly found in group B2 and C, and these strains had a higher resistance to fluoroquinolones, trimethoprim-sulfamethoxazole, and amoxicillin-clavulanate [30]. Among adhesin genes, the prevalence of fimH was the highest (91.8%), followed by pap (79.3%), sfa (12.0%), and afa (7.7%). Additionally, biofilm formation was significantly correlated with the pap adhesion gene [30].A study from Iran reported statistically higher levels of papEF in strong biofilm formers, and the adhesion genes fimH, focG, sfaS, set-1, and cvaC were more common in weaker biofilm formers [31]. We have previously shown that the biofilm forming tendencies of UPEC isolates are variable and not apparent from colony morphology, and that sub-inhibitory concentrations of ciprofloxacin and trimethoprim can, in some cases induce UPEC isolates to form stronger biofilms in vitro, even when resistant to antibiotics [119,120].

2.4. Toxin Production and Cytotoxic Effects

2.4.1. Hemolysin

α-hemolysin (HlyA) is pore forming toxin and virulence factor produced by many strains of UPEC. The prevalence of hemolysin production by UPEC is between 21–47% of isolates, as determined by multiple studies [121,122,123,124]. The operon which encodes α-hemolysin consists of four genes, hlyCABD [125,126]. HlyA can trigger apoptosis in host cells by modulating cell death pathways and interfering with cell process regulators. For example it has been shown that protein kinase B (PBK) which inhibits apoptosis and stimulates immune responses in host cells, is itself inhibited by HlyA in human bladder epithelial cells [127]. The immune response is further disrupted because HlyA inhibits the metabolic enzyme ACLY, which results in low acetyl-CoA levels. This modulates histone acetylation and prevents the adequate promotion of proinflammatory cytokine and chemokine genes [128]. During infection, HlyA stimulates the upregulation of granulocyte-macrophage colony-stimulating factor (GM-CSF) which causes macrophage accumulation and kidney damage during episodes of acute pyelonephritis [34]. UPEC isolates that were found to be stronger biofilm formers in vitro have been shown to have higher hemolysin activity [122].

2.4.2. Cytotoxic Necrotizing Factor Type 1 (CNF1)

Many strains of UPEC produce cytotoxic necrotizing factor type 1 (CNF1), an endotoxin that modulates the activity of host cells by interacting with the Rho family of GTPases and restricting their activation [129]. These GTPases act as molecular switches that control many cellular processes, such as the formation of actin structures, cell motility, cell cycle progression, and phagocytosis [130,131,132]. A range of effects on human and animal cell lines have been documented after exposure to CNF toxins, including elongation and multinucleation, which in vivo will lead to host cell damage and bacterial persistence [35]. There are four known variants of CNF: CNF1, CNF2, CNF3, and CNFY. They each work by deamidating Rho Q63, and the resulting inhibition of GTPase leads to a reorganisation of actin stress fibres, modification of the host cytoskeleton, and cell damage [133,134]. CNF2 differs from the chromosomally mediated CNF1 and CNF3 in that it is plasmid encoded [135].
CNF1 is encoded by the gene cnf1, which has been found in a pathogenicity island that also contains genes encoding P-fimbriae and α-hemolysin [136]. CNF1 internalises into host cells via receptor-mediated endocytosis and contains two distinct binding regions, one located on the N terminus composed of amino acids 135 to 164, and one on the C terminus composed of amino acids 683 to 730, the host receptor being laminin receptor precursor protein (LRP), which is expressed on the surface of eukaryotic cells [137,138].
Epidemiological data suggest that CNF1 is much more common in UPEC as compared to commensal E. coli isolates, and in mouse models it has been shown that CNF1 positive strains more extensively colonise the bladder, are recovered from the urine in greater numbers, and were better able to resist killing by human neutrophils than CNF1 negative strains [139,140]. CNF1 is also associated with E. coli strains that are causative of neonatal meningitis, as well as diarrhoea in children [141,142]. Regarding meningitis, it is thought that E. coli producing CNF1 can more effectively cross the blood–brain barrier and infect the brain, and has been shown to increase disease severity in a mouse model of meningitis [143,144].

2.4.3. Serine Protease Autotransporter

SPATE (serine protease autotransporters of Enterobacteriaceae) are a subgroup of secreted proteins called autotransporters that contribute to the survivability and virulence of UPEC [145]. Among this family, the secreted autotransporter toxin encoded by the sat gene has particular importance to UPEC virulence due to its cytotoxic activity on urinary tract epithelial cells [146]. First discovered in E. coli CFT073, a strain isolated from a patient with acute pyelonephritis, it shows in vitro cytopathic activity in kidney and bladder cell lines and is found significantly more frequently in E. coli associated with pyelonephritis than in faecal E. coli strains [146]. When investigating the kidney damage caused by sat in a mouse model of UTI, Guyer et al. (2002) found that sat induces vacuolation in glomeruli and proximal tubules, a histological change that has been seen in other toxins such as VacA toxin of Helicobacter pylori [147,148]. Sat has also been shown to exhibit immunomodulatory effects, presenting proteolytic effects against complement proteins that would otherwise destroy invading bacteria once they enter the bloodstream following endothelial infection [36].
Table 2 shows virulence factors related to fimbrial and non-fimbrial adhesions with their corresponding genes and host cell receptors, as well as toxins produced by UPEC with associated genes and the corresponding effects on host cells.

2.5. Iron Acquisition Systems

Metal ions are essential nutrients required by UPEC for many crucial biological processes, including the formation of enzymes involved in cellular metabolism [150]. These are metalloenzymes, which require metals including iron, nickel, zinc and copper as co-factors, with iron being the most commonly required [151]. As iron is a key factor in the proliferation of UPEC within the urinary tract, and the retention of iron to deprive invading pathogens of the nutrient composes part of the defensive strategy on the part of the host, acquisition of iron is a key point of competition between host and pathogen [152,153,154]. UPEC have evolved several iron acquisition systems in response to this competition, the first being iron-chelating molecules called siderophores, of which there are four that UPEC can synthesis and secrete, those being enterobactin, salmochelin, yersiniabactin, and aerobactin [155]. Of these, enterobactin is the most conserved among clinical E. coli isolates, with the others appearing in various combinations [155,156]. These variations in genetic carriage of siderophore systems appear to be an important part of the UPEC immune evasion strategy, as host mechanisms can prevent iron acquisition activity of specific siderophores, for example, the innate immune protein lipocalin-2 (Lcn2) prevents enterobactin activity by binding ferric and aferric enterobactin [157]. Enterobactin is encoded by the operon entABCDEF; salmochelin is encoded by the genes iroB and iroE; yersinianactin is encoded by ybtSETU, irp1, and irp2; and aerobactin is encoded by iucABCD. The presence of irp2 and iuc has previously been reported to be most prevalent in serogroups 025, 015 and 08 [52]. Rezatofighi et al. (2021) has reported that siderophores genes fyuA and iutA are found more frequently in group B2 than in other phylogroups [158] While iutD has been reported to be highly distributed into groups B2 and D among multidrug-resistant UPEC (MDR-UPEC) and extensively drug resistant UPEC (XDR-UPEC) [118]. In addition to siderophores, UPEC also utilise host haemoglobin to acquire iron using the outer membrane receptors ChuA and Hma [159,160]. Strains lacking either of these receptors has been shown to be ineffective colonisers of the kidneys in a mouse model of UTI, indicating their importance for successful colonisation of the upper urinary tract [160]. Even among asymptomatic bacteriuria strains, which lack many of the common virulence factors expressed by other UPEC, they will express iron acquisition systems, showing their necessity in urinary tract colonisation [161].
Fimbrial adhesions, curli, and non fimbrial surface adhesions are expressed on the cell surface to aid in colonisation and persistence through UPEC and host cell interactions, facilitating intra-cellular invasion as well as cell aggregation and biofilm formation. The expression of these surface proteins is regulated by phase variation. Outer membrane receptors and siderophores sequester iron which is essential for many cell processes and a scarce resource in the urinary tract. Secreted toxins cause tissue damage in an infected host and assist in immune evasion.

3. Antibiotic Resistance Mechanisms

Antibiotic resistance mechanisms in UPEC can include chromosomal mutations in target genes and efflux pumps which allow for enhanced expulsion of antimicrobials from the cell interior. Additionally, enzymes can be produced which either deactivate the antimicrobial by degradation or by competing competitively for bindings sites. Here, the major mechanisms of resistance to clinically relevant antibiotics used in the treatment of UPEC-associated UTIs are described. These mechanisms are summarised in Table 3, and the cell targets of antimicrobials used in the treatment of UPEC associated UTIs are shown in Figure 2.

3.1. β-Lactams

β-lactams are a wide category of antibiotics that include both wide and narrow spectrum penicillin, cephalosporins (first, second, third, fourth and fifth generation), monobactams, and carbapenems. Clinically, there are five classes: penams, cephems, carbapenems, clavams, and monobactams. They produce a bactericidal effect by binding to penicillin-binding proteins and inhibiting bacterial cell-wall formation [162]. Resistance to cephalosporins in UPEC can occur through various mechanisms, including β-lactamase production, target site mutations, and efflux pumps [163,164,165].
β-lactam resistance in UPEC is primarily due to the production of β-lactamases, enzymes that cleave the integral β-lactam ring and render these antibiotics ineffective. The most common type of β-lactamase produced by UPEC is CTX-M-type extended-spectrum β-lactamase (ESBL), which hydrolyses third generation cephalosporins and monobactams as well as other β-lactams [166]. A recent study by Mohammed et al. (2022) found that ESBL production was positively associated with certain serotypes, namely O8, O15, O21, O22, and O25 while being negatively associated with serogroups O1, O4, O7, and O16 [53]. Additionally, ampicillin resistance has been found to be significantly higher among serogroups O15 and O22 [10]. Molecular analysis of UPEC in Iran found that blaCTX-M and blaTEM genes were found in 53.2% and 45% of UPEC respectably, whereas the blaSHV gene was less commonly found at 5.4% [163]. These β-lactamase genes are often found on plasmids that can be easily transferred between bacterial strains that carry with them aminoglycoside, dhfr and sul genes. Additionally, a positive correlation has been observed between ESBL producing UPEC and the presence of biofilm formation genes pgaA and pgaC [167]. AmpC β-lactamase can confer resistance to aminopenicillins, cephalosporins, and monobactams, and they are not affected by clavulanic acid, an ESBL inhibitor [168]. In E. coli, AmpC production can be mediated by plasmids carrying ampC genes (pAmpC) [169], but also through chromosomal mutations. In this case, the promoter/attenuator region of the chromosomal cephalosporinase gene (ampC) is mutated, resulting in overexpression [170]. Carbapenems are antibiotics of last resort and the emergence of carbapenem-resistant UPEC (CR-UPEC) is an internationally recognised societal challenge [171]. The most common carbapenemase found in UPEC is the New Delhi metallo-β-lactamase (NDM), encoded by the blaNDM gene, which has been found in globally distributed UPEC isolates [172].
In addition to β-lactamases, UPEC can also acquire resistance through mutations in genes involved in cell wall synthesis. For example, mutations in the penicillin-binding proteins (PBPs) can lead to a decrease in the affinity of β-lactams for their targets, thereby decreasing their efficacy. Studies have characterised mutations in mrdA, the gene encoding penicillin binding protein 2, finding ten different patterns which increased in MIC of carbapenems [164] Mutations in mrdA have also been implicated in reduced activity of diazabicyclooctane, a B-lactamase inhibitor [173]. Efflux pumps are another mechanism by which UPEC can develop resistance to cephalosporins. The efflux pumps AcrAB-TolC and AcrAD-TolC have been shown to be over expressed in carbapenem resistant E. coli. One study found a strong relationship between AcrA over expression and ertapenem resistance, and AcrB over expression due to imipenem induced stress in E. coli [165]
Recent years have seen the introduction of novel β-lactam/β-lactamase inhibitor combinations for use against Gram-negative bacteria. Namely, ceftolozane/tazobactam and ceftazidime/avibactam. Both drugs combine a 2nd generation cephalosporin with an inhibitor which targets the active site of serine β-lactamases [174]. Both inhibitors, tazobactam and avibactam, effectively inhibit the activity of TEM-1, TEM-2, TEM-3, SHV-1, SHV-2, and CTX-M-14, However, neither will inhibit class B metallo-β-lactamases, such as NDM-1, and only avibactam shows effective inhibition of AmpC β-lactamase and Klebsiella pneumoniae carbapenemase (KPC) [174,175] This gives both antibiotic-inhibitor combinations a wide spectrum of activity against ESBL-producing UPEC, and meta-analysis of randomised control trials have indicated that the use of these drugs results in better outcomes for the treatment of complicated UTI when compared to alternative antibiotic options [176]. The main mechanisms of resistance for both drugs in UPEC are class B metallo-β-lactamases; however, other mechanisms that are not enzymatically mediated can still impart resistance, for example, mutations in PBP3, the target site for ceftazidime has been shown to render E. coli resistant to ceftazidime-avibactam [177]. Likewise, the efflux pump AcrAB has been demonstrated to increase the MIC of ceftolozane/tazobactam to Enterobacteriaceae previously [178].

3.2. Nitrofurantoin

Nitrofurantoin remains an effective first-line treatment and prophylaxis for lower UTIs due to the low resistance rates observed [179]. The slower development of resistance to nitrofurantoin compared with other antimicrobials may be due to the unique and incompletely understood mode of action. Bacteria first catalyse the breakdown of nitrofurantoin to several bioactive metabolites that possess antimicrobial properties using the bacterial oxygen-insensitive nitroreductases enzymes, expressed by the nfsA and nfsB genes [179,180]. The metabolites achieve their bacteriostatic affect by binding to bacterial ribosomes and inhibiting protein synthesis, as well as by inhibiting enzymes involved in the citric acid cycle and those involved in the synthesis of DNA and RNA [181]. The main mechanism of resistance to nitrofurantoin are deletion and insertion mutations in nfsA and nfsB genes. Additionally, resistance can be conferred due to mutations in the ribE gene which encodes for lumazine synthase, an enzyme which has involvement in the synthesis of the NfsA and NfsB co-factor flavin mononucleotide [182,183].
Resistance to nitrofurantoin can also be plasmid mediated. Firstly, the multidrug-efflux gene oqxAB is carried by a variety of plasmids types, and is commonly flanked by IS26 comprising the transposon Tn6010 [184]. While it remains unclear whether or not high-level resistance can develop from carrying the oqxAB gene alone, high level nitrofurantoin resistance results from nfsA inactivation combined with expression of the oqxAB gene [185]. Nitrofurantoin-resistant strains in fact show a higher prevalence of efflux genes than other resistant phenotypes, and 51% of them have been shown to be extensively drug resistant (XDR), exhibiting co-resistance to β-lactams, cephalosporins, carbapenems, aminoglycosides, and tetracyclines [186].
Recently, in a Welsh hospital, a second plasmid-mediated resistance mechanism was discovered—a mutated form of CTX-M-14 β-lactamase enzyme isolated from five UTI-positive urines. Three specific associated mutations (T55A, A273P, and R277C) were found to both significantly increase the minimum inhibitory concentration (MIC) to nitrofurantoin when recombinantly expressed in E. coli (Bio-85025) and to be capable of hydrolysing nitrofurantoin in a cell-free assay [187]. Potential dissemination of this gene, via horizontal gene transfer or clonal expansion, is an emerging clinical consideration.

3.3. Fosfomycin

Like nitrofurantoin, the resistance rates to fosfomycin among UPEC remains low, preserving its clinical utility in the treatment of UTIs [179,188]. The mechanism of action is also unique, meaning that the development of co-resistance is unlikely [189]. That said, resistance is increasing, especially among extended-spectrum β-lactamase (ESBL) producers [190]. Fosfomycin acts as a competitive inhibitor of the enzyme UDP-N-acetylglucosamine-enolpyruvyltransferase (MurA), which catalyses peptidoglycan synthesis in E. coli, by binding to the cysteine 115 residue [191]. Because of this, a C115D MurA mutant leads to ineffective inhibition and associated fosfomycin resistance [192].
For inhibition to occur, fosfomycin requires intracellular transport facilitated by the glycerol-30-phosphate (G-3-P) transport system (glpT) as well as the hexose phosphate uptake transport system (uhpT) [193]. Mutations in these transport genes have been shown to increase the MIC of E. coli for fosfomycin, but not to the degree where it is outside EUCAST susceptibility range, indicating that a multifactorial series of mutations and deletions in combination with other resistance mechanisms are required for fosfomycin resistance to develop through chromosomal gene mutations alone [194].
A series of plasmid-encoded metalloenzymes, fos genes, confer resistance by modifying fosfomycin to an inactive form, with the most common found in E. coli being fosA3 [191]. A study in Mexico which determined the mechanism of resistance to fosfomycin among 350 ESBL-producing E. coli found that among the fosfomycin-resistant strains, 60.5% of them were fos-producing, with the majority of those strains carrying fosA3, while resistance due to mutations in the antibiotic transport system was responsible for 28.9% of resistant strains [195]. Recently, fosA8 coding a protein with 80% identity to fosA, was identified from an E. coli isolated from urine, located on a transferrable plasmid and conferring high-level resistance [196]. This gene was traced to the genome of Leclercia decarboxylata, a species with natural fosfomycin resistance and a reservoir of transferable resistance genes [196]. Another plasmid mediated resistance gene, named fosL1, was identified from isolates in Switzerland and believed to be acquired by Tn7-related transposition. This gene shared 57% to 63% amino acid identity with other fosA genes [197].
Fosfomycin is available for use in both oral and intravenous formulations. Oral fosfomycin can be administered in the form of fosfomycin trometamol and fosfomycin calcium, while intravenous fosfomycin is formulated with disodium in a ratio of 1 g to 8 g with succinic acid as an excipient [198]. For oral formulations, the maximum concentration is reached in the urine after 2 h and will maintain a mean concentration above 128 mg/L for 36 h after administration with a 3 g dose [199]. While oral formulations are recommended as a first-line treatment for uncomplicated UTIs in women with infections caused by E. coli, is has also been shown to be effective in treating male UTIs and acute bacterial prostatitis [7,200]. This is due to the fact that, apart from the urine, elevated intraprostatic concentrations are also reached following administration [201].
Intravenous doses of fosfomycin, on the other hand, have a faster period of absorption and the urinary concentration will drop below 128 mg/L 12 h post administration [199]. The maximum plasma concentrations reached through IV administration are much higher, however, it has been shown that significant concentrations are reached at various body sites [199,202]. The favourable PK/PD profile coupled with the unique mode of action of fosfomycin, means that IV fosfomycin is particularly useful as a combination therapy with other antimicrobials. For example, it has been shown to be effective when used in combination with cefiderocol, a siderophore antibiotic resistant to several β-lactamases, including metallo-β-lactamases, to treat MDR Gram-negative bacteraemia in patients with impaired renal function [203].

3.4. Trimethoprim-Sulfamethoxazole

Trimethoprim inhibits the formation of tetrahydrofolic acid, a cofactor required for the successful synthesis of bacterial DNA. It is commonly paired with the sulphonamide drug sulfamethoxazole, which inhibits the synthesis of dihydrofolic acid [204]. Resistance to trimethoprim and sulfamethoxazole is highly associated with mobile genetic elements, plasmids, and integrons [205,206]. The genes which encode these resistance proteins have their ancestral roots in chromosomal folA and folP, from a species which have a natural resistance to trimethoprim and sulphonamides [207]. Dfr (di-hydrofolate reductase) are a large family of genes conferring resistance to trimethoprim. There are over 30 dhfr genes, with the most common in UPEC being dfrA1 and dfrA17 [208]. These dfrA genes are often found in close genomic proximity to sul genes—which confer resistance to sulfamethoxazole—within integrons and on plasmids. They are also commonly found among larger resistance islands that often contain blaCTX-M, chromate-resistance gene chrA, aminoglycoside genes aac(3)-IIa, aac(6)-Ib-cr, aph(3′)-Ia, and aadA2 as well as other virulence genes fimH, fyuA, irp2, and sitA [209]. The inherent capacity for mobility associated with these resistance determinants has caused extensive spread of trimethoprim-sulfamethoxazole resistance among UPEC [205,210,211]. Selective pressure presented by previous antibiotic therapy has been shown to lead to the trimethoprim resistance, specifically the use of extended-spectrum penicillin, however, previous prescription with nitrofurantoin was found to be associated with a lower incidence of trimethoprim resistance [212].
Although these mobile determinants play the largest role in trimethoprim-sulfamethoxazole resistance, both efflux and chromosomal mutations have also been demonstrated to contribute to resistance to these agents. Regarding efflux, the AcrAB-TolC efflux system leads to increased trimethoprim MICs in E. coli and was shown to be upregulated due to the effects of decreased periplasmic glutathione concentration caused by loss-of-function mutations of the genes gshA, grxA, and cydD—known to regulate GSH content [213]. This leads to the speculation that mutations in these genes could mediate resistance to trimethoprim chromosomally. Furthermore, a study which screened a knockout library of E. coli found that mgrB, when deleted, resulted in trimethoprim resistance. phoP, phoQ, and folA were significantly upregulated in mgrB-deleted strains. This is because mgrB regulates the expression of folA, showing that many genes involved in folate metabolism can lead to trimethoprim resistance [214].

3.5. Fluoroquinolones

Ciprofloxacin, levofloxacin, norfloxacin, ofloxacin, and gatifloxacin are examples of fluoroquinolones used to treat UTI, and can have broad spectrum efficacy against both Gram-negative and Gram-positive bacteria [215]. The EAU does not recommend ciprofloxacin for uncomplicated UTI but recommends its use for cases of complicated UTI where the local resistance rates are below 10% and the patient has contraindications for third-generation cephalosporins or an aminoglycoside [7].
Fluroquinolones prevent bacterial DNA synthesis by inhibiting DNA gyrase and topoisomerase IV. DNA gyrase is responsible for introducing the negative supercoiling of double stranded DNA, while topoisomerase IV disentangles replicated DNA and allows for segregation of daughter chromosomes. DNA gyrase is composed of two subunits, GryA and GyrB, while topoisomerase is composed of four subunits, two ParC subunits and Two ParE subunits [216]. Chromosomal mutations to these targets, typically GryA, result in resistance to fluroquinolones. In a study of fluroquinolone-resistant E. coli, Esmaeel et al. (2020) found that 76.7% had mutations in the GyrA subunit, and the highest levels of resistance were associated with strains that possessed a dual Ser-83 and Asp-87 mutation in that subunit [217].
Resistance to fluoroquinolones is also highly dependent on the ability of bacteria to decrease drug accumulation by efflux, though different efflux systems vary in their contribution to resistance for different fluroquinolones. For instance, AcrB expression is significantly higher in ciprofloxacin-resistant E. coli, 26% higher than in susceptible isolates [218], although it is unlikely to be the predominant mode of fluoroquinolone resistance. Specific quinolone efflux pump genes qepA and oqxAB as well as plasmid-mediated quinolone resistance (PMQR) genes, which include qnrA, qnrB, qnrC, qnrS, qnrD, qnrE, and qnrVC are plasmid-encoded and can provide resistance in E. coli even in the absence of chromosomal mutations [219]. A study which looked at the distribution of qnr genes among ESBL-producing E. coli found that qnrB was present in 47.74%, qnrS was found in 47.10%, and qnrA was found in 2.58% of strains. These genes were highly associated with the ESBL genes blaCTX-M, blaTEM, blaSHV [220]. Another study found qnrS in 21 of 43 isolates, qnrB in 6 of 45 isolates and qnrA in 2 of 43 isolates [221]. They are highly transmissible and are transferred in tandem with additional β-lactams, gentamicin, and tetracycline resistance genes [222].
The most commonly reported PMQR mechanism is the AAC(6′)-Ib-cr enzyme which dually inactivates ciprofloxacin while additionally providing aminoglycoside resistance. It has been found to be present in >70% of strains from the subgroup sequence type ST-131, an antibiotic resistant clone that has been reported to be rapidly spreading and responsible for high rates of UTI and bloodstream infections [223]. While the enzyme alone imparts a low-level MIC increase for ciprofloxacin in E. coli, it enhances resistance when combined with resistance-associated chromosomal mutations [224].
DNA synthesis, protein synthesis, peptidoglycan synthesis, cell membrane synthesis, and folic acid synthesis can be inhibited by targeting key molecules using different antibiotic agents.
Table 3. Chromosomal, enzymatic, and efflux mediated resistance mechanisms commonly found in E. coli contributing to antibiotic resistance.
Table 3. Chromosomal, enzymatic, and efflux mediated resistance mechanisms commonly found in E. coli contributing to antibiotic resistance.
Chromosomal MutationsEnzymatic Mechanism (Plasmid Encoded) Efflux Mediated
NitrofurantoinnfsA, nfsB [182] ribE [183]CTX-M-14 (mutated) [187]oqxAB [185]
FosfomycinmurA, [192]
glpT, uhpT [194]
fosA, fosL [195,196,197,225]
(Various alleles)
Trimethoprim mgrB [214]dfrA1, dfrA5, dfrA7, dfrA12, dfrA17 [226]
dfrA8, dfrA14, dfr2d, dfrA3, dfrA9, dfrA10, dfrA24, dfrA26 [227]
acrAB-tolC [213]
Sulfamethoxazole sul1, sul2, sul3 [228]
FluoroquinolonesgryA, gyrB, parC [217]qnrA, qnrB, qnrC, qnrS, qnrD, qnrE, qnrVC [219]
aac(6′)-Ib-cr [223]
acrB, [218]
qepA, oqxAB [219]
β-lactamsmrdA [164]
ampC [92]
blaCTX-M, blaTEM, blaSHV [163],
blaOXA, blaCMY [229]
pAmpC [91]
acrAB-tolC, acrAD-tolC [165]
Table 3 shows resistant determinants of note for each antibiotic/antibiotic class stemming from either chromosomal gene mutation/deletion, antibiotic inactivation due to an enzymatic mechanism, or by efflux.

4. The Mobilome

4.1. Plasmids

Plasmids, which are extrachromosomal DNA molecules, can be classified and typed using various methods based on replicons [230], partition systems [231], mob genes for relaxases [232], and comparison of profiles generated through plasmid digestion with restriction enzymes [233]. Sub-typing can be used to further differentiate plasmids by double locus sequence typing [233]. The most commonly used typing method today is Inc/rep typing, which classifies plasmids based on their ability to stably coexist with other plasmids in the same bacterial strain—facilitated by the replication machinery within the plasmid [234].
Epidemic plasmids, such as IncF, IncI, IncA/C, and IncH, are the most abundant types of plasmids [235]. Among UPEC specifically, IncFIB, and IncB/O plasmid replicons are the most common [236]. However, the prevalence of certain plasmids differs depending on their sources and geographic locations [235]. These plasmid classes also appear to have different correlations with different antibiotic resistance genes, for example Incl1 plasmids are more often associated with clinically relevant strains and likely possess genes encoding ESBLs and AmpC enzymes [237]. More recently, the plasmid encoded colistin resistance gene mcr-1 (phosphoethanolamine transferase) was detected on E. coli Incl1 plasmids recovered from Canada and Argentina [238]. IncF plasmids differ in that they can encode multiple replicons, and can carry blaCTX-M, rmtB, oqxB as well as qnr genes [239]. IncB/O plasmids, while less common, carry a great variety of resistance genes, including blaCTX-M−1, blaCMY−2, blaACC−4, blaSCO−1, blaTEM−1, sul1, sul2, aad, strA, strB and aacA4 [235].
Plasmids carry genes encoding resistance to nitrofurantoin [185] β-lactams [240], fluoroquinolones [222], aminoglycosides [241], trimethoprim [209], sulfamethoxazole [242], tetracyclines [243], and fosfomycin [195] among others. These genes are often distributed within plasmids in different combinations, allowing for the spread of multidrug resistance during horizontal transfer events [66].
A study which looked to determine the distribution of PMQR determinants among ciprofloxacin resistant UPEC isolates, as well as co-occurrence of ESBL production and β-lactamase inhibitor resistance, found that 50.0% of the isolates carried PMQR determinants, with ESBL production observed in 42.9% of these isolates and a β-lactamase inhibitor-resistant phenotype in 51.4%. Furthermore, co-occurrence and co-transmission of aac(6′)-Ib-cr with blaTEM, blaCTX-M, and blaOXA was the most prevalent, observed in 37.1% of the isolates [244]. An Egyptian study on the distribution of PMQR in UPEC found that the aac(6′)-Ib-cr gene was the most common (61.1%), followed by qnrS (43.3%) and qnrB (22.2%). The qepA gene had a low frequency (10%), while qnrA was not detected. The aac(6′)-Ib-cr gene was found to exist either alone or in combination with qnrS and/or qnrB. Interestingly, the coexistence of qnrS and qnrB genes was observed in only six isolates [217]. Another study characterised the plasmids of 25 UPEC each carrying one to two plasmids per strain and found that combinations of genes encoding β-lactamase, aminoglycoside modifying enzymes, dihydrofolate reductase, dihydropteroate synthase, and macrolide 2′ phosphotransferase were present in plasmids belonging to seven incompatibility groups, with IncFI and IncFII being the predominant resistance plasmids [66].
Studies have shown that plasmids can transfer between E. coli strains in a biofilm, and some plasmids have consistently higher transfer efficiency in biofilms when compared to liquid culture [32]. Another study focused on the effect of oxygen on IncP-1 plasmid transfer efficiency in biofilms, finding that the air-liquid interface can be a hot-spot for plasmid mediated gene transfer due to a high juxtaposition of donor and recipient cells [245]. The ability of plasmids to conjugate was also found to be dependent on biofilm biomass, with thinner biofilms allowing for increased frequency of conjugation. This suggests that the development of multidrug-resistant UPEC might be correlated with lower biofilm formation due to being a good recipient in conjugative transfer [246].

4.2. Integrons

Integrons are a mobile genetic element which assists in the horizontal transfer of multidrug-resistance genes in UPEC. Class 1 integrons can capture and express gene cassettes through a promoter (Pc) and primary recombination site. In addition, integrons can relocate within the host genome and other cells through their association with transposons and plasmids [247]. Integrons are common in E. coli, with one Chinese study reporting that 67.39% (93/138) of E. coli isolates carried class 1 integrons. Integrons have also been reported to be more common in UPEC than in commensal E.coli isolates [248].
The most common gene cassette found is dfr and aadA, conferring resistance to trimethoprim, streptomycin, and spectinomycin [247]. Other studies have indicated that there is a strong correlation between multi drug resistance and the presence of a class 1 integrons in UPEC [249,250,251]. A Mexican study which reported 63% of E. coli strains possessing the ilt1 gene also found that the phylogenetic group B2 were more likely to have a class 1 integron. They went on to report the diversity of the cassettes finding eight variations with the dfrA17 and aadA5 arrangement present in 32.1% of variable regions [250]. Phylogenetic groups B2, A, and D have also been reported to harbour class 1 and 2 integrons in MDR-UPEC and XDR-UPEC, carrying the resistance genes aadA1, aadB, aacC, ant1, dfrA1, dfrA17, and aadA4 [118]. Expression studies have shown a strong correlation between class 1 integrons and resistance to trimethoprim and cefotaxime [205].
The is also a strong correlation between Int1 integrase and sulphonamide resistance, shown in one study were the authors examined 50 strains positive for the IntI1, and found that all of these strains were resistant to sulfamethoxazole, with the majority containing sul1, either alone or with sul2 or sul3 [252].

4.3. Transposons and Insertion Sequences

Transposons and insertion sequences are further examples of mobile genetic elements that contribute to antimicrobial resistance in UPEC [228]. They do this by moving to new locations within a genome, or transferring from one genome to another, causing genetic rearrangements, interrupting genes, and carrying gene cassettes containing antibiotic resistance genes and virulence factors to plasmids, allowing for the horizontal spread of these genes to different bacterial species [253].
Insertion sequences are common in E. coli, with two or three per isolate and IS26 being the most common [254]. One study investigated the role of transposable elements in E. coli BH100, a strain closely related to UPEC strains, finding that IS4 and IS21 were associated with genomic islands containing genes associated with antimicrobial resistance and virulence. Additionally, the study found that a TN3 transposon harbouring β-lactamase TEM-1 was transmitted between plasmids of different replicon type [255]. Recombination events involving IS26 elements have been shown to impart the qnr resistance gene qnrB19 to an E. coli plasmid isolated from animal origin [256].
The interruption of nfsA and nfsB genes was reported in UPEC studied in the United Kingdom, leading to nitrofurantoin resistance due to gene inactivation by the insertion sequences IS1 and IS10R, respectively [182]. Additionally, insertion sequences and transposons also play a role in the formation and evolution of cointegrate plasmids, which are plasmids formed by the fusion of two or more plasmids that share homologous sequences and can promote the dissemination of multidrug resistance. For example, IS1294 has previously been shown to mediate fusion between plasmids with different replicons, facilitating the co-transfer of blaCTX-M−55 and rmtB [257].

5. Perspective Alternative Treatments for UTI

Developing and utilizing alternative therapies such as phage therapy, immunomodulation, vaccines, and novel antimicrobial agents offers a multifaceted approach to combat the challenge of escalating antibiotic resistance in UPEC. These alternatives can provide targeted, effective treatments for infections that are refractory to antibiotics, reduce the risk of further resistance development, and mitigate the adverse effects associated with antibiotic use. Presented here is an overview of potential novel treatments for UTI.

5.1. Vaccines and Immunomodulation Therapies

The development of vaccines for preventing UTIs is a dynamic and evolving field and recent studies have explored diverse strategies to harness the immune system’s ability to protect against UTIs. Previously, studies have investigated and found siderophores and heme iron receptors as promising candidates for antigens as part of a vaccine formulation [258,259]. In 2015, four antigens associated with iron acquisition (IreA, Hma, IutA, FyuA) were investigated and were found to generate a strong antigen-specific serum IgG which protected vaccinated mice, and demonstrated antibody binding to UPEC strains expressing those antigens [260]. Later in 2020, steps towards the optimisation of a vaccine using these antigens were made, and it was then was found that it provided the best protection against UPEC UTI when formulated with dmLT adjuvant [261]. Also, that year, J. Wu et al. investigated the use of intravesical vaccination with UPEC antigens and a Th1-skewing adjuvant to correct aberrant bladder immune responses. Intravesical immunization demonstrated marked protection against UTIs, attributed to its distinctive ability to recruit Th1 cells into the bladder. This approach proved effective even in mice with multiple UTIs and pronounced abnormal immune responses [262]. In 2022, a report from a multicentre randomised trial conducted by Lorenzo-Gómez et al. evaluated a sublingual bacterial preparation called MV140 for preventing recurrent UTIs in women. MV140 was found to reduce the incidence of positive urine cultures across bacterial species. Notably, the protection conferred by MV140 was durable for at least one year, setting it apart from short-term treatments, like antibiotic prophylaxis, that exhibit rapid loss of efficacy post-discontinuation [263]. Also in 2022, Kelly et al. introduced a supramolecular peptide nanofiber vaccine platform designed to elicit specific immune responses against UPEC. By assembling multiple pathogen-specific epitopes into nanofibers, the platform successfully generated robust anti-UPEC antibodies in mice. The resulting antibodies were effective against UPEC and demonstrated both systemic and mucosal presence. Furthermore, the nanofiber vaccine caused minimal disruption to the gut microbiota, a favourable aspect compared to antibiotics. Collectively, these recent studies showcase a range of innovative strategies in the development of UTI vaccines [264].
Aside from vaccines, there has been reported successes in using alternative immunomodulation therapies. These therapies aim to either stimulate or suppress specific components of the innate immune system to achieve a balanced and targeted response against bacterial infections [265]. These therapies target specific disease-related-immune-response regulators and have been demonstrated to be efficacious at controlling disease through modulating inflammation leading to more effective bacterial clearance from the kidneys and bladder in animal studies [265]. For example, inhibition of transcription factor interferon regulatory factor 7 (IRF-7) through the use of siRNAs was found to protected against infection and renal tissue damage in mice with pyelonephritis [266]. By the same token, the cytokine Interleukin-1 beta (IL-1β) has been found to be a key immune regulator in acute cystitis, and its suppression using IL-1RA anakinra as an inhibitor attenuated inflammation in infected mice, resulting in accelerated bacterial clearance [267].

5.2. Phage-Based Therapeutics

Phage-based therapeutics have several enticing advantages over antibiotics. For one, they have a comparatively lower potential for the development of resistance, owing to the fact that phages typically have a narrow host range, are strictly bactericidal, and mutational changes to phage receptors that cause resistance are also likely to impact fitness and virulence of bacteria [268,269]. Additionally, these mutations can lend itself towards increased sensitivity to antibiotics, for example, Phage U136B requires genes involved in the formation of LPS (rfaC, rfaD, rfaE, and rfaP) to successfully invade a host cell [270]. Mutations in these genes result in truncated LPS, and hyper sensitivity to several antibiotics [271]. Phages are also effective at controlling biofilms, especially when used in combination with other agents [272]. A particular utility for bacteriophages as an anti-biofilm coating for urinary catheters has been demonstrated previously [273]. For these reasons, there has been notable research interest in the development of a phage-based therapy for UTIs and there are many reported in vitro and animal studies on the use of bacteriophages in this regard [274,275,276,277]. Clinical studies, too, show promise. Recent reports include a case study reporting the successful treatment of recurrent ESBL Klebsiella pneumoniae urinary tract infection with the use of intravenous phage as a standalone therapy [278]. However, additional robust randomised clinical trials are required to properly assess the efficacy and safety of phage therapy and will be crucial to form a roadmap towards integrating phage therapy with traditional clinical practice [279].

5.3. Antipathogenic Agents

Antipathogenic agents can be used as part of an overall strategy to mitigate the potential to cause disease in UPEC, by interfering with certain virulence pathways, and in a sense, disarm virulent bacteria rather than interrupt their growth in the way antibiotics do. In this way, the selection for antibiotic resistance can be avoided [280]. For example, small molecule compounds have been developed to target type 1, P type, and S type adhesive fimbriae in UPEC [281]. These molecules are called pilicides, and can either directly prevent the assembly of the fimbria structure by binding to the associated chaperone proteins, or by inhibiting the transcription of fimbria genes [282]. Curlicides are small molecules which inhibit curli biogenesis, and include FN075, which was found to inhibit the assembly of type 1 as well as prevent the biogenesis of curli in UPEC, resulting in a less virulent and biofilm deficient strain following attenuation in a mouse model of UTI [283]. Quorum sensing is another virulent pathway which may be interrupted for therapeutic benefits, and a large number of compounds, including plant extracts, have been found which can reduce motility in bacteria, inhibit biofilm formation, or make biofilms more susceptible to antibiotics by affecting quorum sensing [284,285]. While these compounds have shown promising results to limit the pathogenic potential of UPEC in vitro, they are yet to be developed to the point where they can be formulated as a therapeutic.

5.4. Nano Formulations

The application of nanotechnology to drug formulations can enhance the properties of conventional drugs, increasing their absorptivity, their specificity for target sites, and allowing for them to be attached to drug delivery systems [286,287]. Drugs formulated this way are typically 0 to 100 nm and possess unique features, such as a high surface to mass ratio, increased absorptivity, bioavailability, retention time, and reduced side effects over traditional macro formulations [286]. The benefit to antibiotic therapy is that, with nano-technology, the solubility and gastrointestinal contact area increases for oral formulations, and a higher uptake can be achieved through controlled and timed release of the bioactive ingredients [288,289]. Of particular interest, nano preparations could be useful in overcoming resistance mechanisms utilized by bacteria [290]. Antibiotics which are formulated smaller, or have appropriate materials incorporated with them can more easily transverse cellular membranes and access cellular components, and through massively increased drug accumulation within bacterial cells can overcome efflux mechanisms [291], [292]. They can also be formulated to exhibit multiple and varied modes of action, to make the development of resistance more unlikely as compared to the use of antibiotic in its molecular form [293]. Nanoparticle antimicrobials based on metallics (gold, silver, titanium, and zinc) have intrinsic antimicrobial properties and additionally are effective at eradicating biofilms, making them good potential candidates for the treatment of UTIs [294]. These metallic nanoparticles can also be used as drug carriers for existing antibiotics [291,295]. Delafloxacin-gold nano-formulation, a fluoroquinolone antibiotic using gold nanoparticles as a delivery vehicle was found to be more effective against Gram-negative and Gram-positive strains than delafloxacin alone, even when used at lower concentrations [295], and a ciprofloxacin-gold formulation has been found to overcome resistance mediated by efflux in multidrug-resistant Pseudomonas aeruginosa and E. coli [291]. That being said, the interactions between these metals and human tissues, as well as the effect of long-term exposure, requires further in vivo study.

6. Conclusions

UTIs caused by UPEC continue to have a significant impact on the global disease burden, and clinical treatment is considerably complicated by the development and spread of virulence factors and multidrug resistance. Resistance to most clinically useful antibiotics is often multifactorial, stemming from genomic mutations, inactivating enzymes harboured on extrachromosomal plasmids, and drug efflux systems. This is further aided by the genomic plasticity of the pathogen, which includes various mobile genetic elements such as plasmids, transposons, insertion sequences, and integrons. In order for clinical treatment to remain effective in the face of ever-increasing multi-drug resistance in UPEC, continued elucidation of the molecular factors which underpin resistance is necessary.

Author Contributions

Conceptualization, S.W. and B.L.; formal analysis, S.W.; writing—original draft preparation, S.W.; writing—review and editing, B.L. and K.F.; supervision, B.L. and K.F.; project administration, B.L.; funding acquisition, B.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the RISAM scholarship from Munster Technological University [RISAM_SW_2018].

Acknowledgments

The authors would like to acknowledge Caoimhe Lynch for providing independent guidance and proofreading this manuscript. Figure 1 and Figure 2 were created with BioRender.com. Accessed on 18 August 2023.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Yang, X.; Chen, H.; Zheng, Y.; Qu, S.; Wang, H.; Yi, F. Disease burden and long-term trends of urinary tract infections: A worldwide report. Front. Public Health 2022, 10, 888205. [Google Scholar] [CrossRef]
  2. Foxman, B.; Brown, P. Epidemiology of urinary tract infections: Transmission and risk factors, incidence, and costs. Infect. Dis. Clin. N. Am. 2003, 17, 227–241. [Google Scholar] [CrossRef]
  3. Rowe, T.A.; Juthani-Mehta, M. Urinary tract infection in older adults. Aging Health 2013, 9, 519–528. [Google Scholar] [CrossRef]
  4. Öztürk, R.; Murt, A. Epidemiology of urological infections: A global burden. World J. Urol. 2020, 38, 2669–2679. [Google Scholar] [CrossRef]
  5. Vallejo-Torres, L.; Pujol, M.; Shaw, E.; Wiegand, I.; Vigo, J.M.; Stoddart, M.; Grier, S.; Gibbs, J.; Vank, C.; Cuperus, N.; et al. Cost of hospitalised patients due to complicated urinary tract infections: A retrospective observational study in countries with high prevalence of multidrug-resistant Gram-negative bacteria: The combacte-magnet, rescuing study. BMJ Open 2018, 8, e020251. [Google Scholar] [CrossRef]
  6. Callan, A.; O’Shea, E.; Galvin, S.; Duane, S.; Corry, O.; SIMPle Team, T.; Vellinga, A. The Economic Cost Of Urinary Tract Infections In The Community: Results From Ireland. Value Health 2014, 17, A468. [Google Scholar] [CrossRef]
  7. Bonkat, G.; Bartoletti, R.; Bruyère, F.; Cai, T.; Geerlings, S.E.; Köves, B.; Schubert, S.; Pilatz, A.; Veeratterapillay, R.; Wagenlehner, F. EAU Guidelines on Urological Infections. Eur. Assoc. Urol. 2023, 182, 237–257. [Google Scholar]
  8. Aggarwal, N.; Lotfollahzadeh, S. Recurrent Urinary Tract Infections; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
  9. Nazarko, L. Recurrent urinary tract infection in older women. NursePrescribing 2014, 12, 608–613. [Google Scholar] [CrossRef]
  10. Medina, M.; Castillo-Pino, E. An introduction to the epidemiology and burden of urinary tract infections. Ther. Adv. Urol. 2019, 11, 1756287219832172. [Google Scholar] [CrossRef]
  11. Kaper, J.B.; Nataro, J.P.; Mobley, H.L.T. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004, 2, 123–140. [Google Scholar] [CrossRef]
  12. Manges, A.R.; Geum, H.M.; Guo, A.; Edens, T.J.; Fibke, C.D.; Pitout, J.D.D. Global Extraintestinal Pathogenic Escherichia coli (ExPEC) Lineages. Clin. Microbiol. Rev. 2019, 32. [Google Scholar] [CrossRef]
  13. Gomes, T.A.T.; Elias, W.P.; Scaletsky, I.C.A.; Guth, B.E.C.; Rodrigues, J.F.; Piazza, R.M.F.; Ferreira, L.C.S.; Martinez, M.B. Diarrheagenic Escherichia coli. Braz. J. Microbiol. 2016, 47 (Suppl. 1), 3–30. [Google Scholar] [CrossRef]
  14. Akira, S.; Takeda, K.; Kaisho, T. Toll-like receptors: Critical proteins linking innate and acquired immunity. Nat. Immunol. 2001, 2, 675–680. [Google Scholar] [CrossRef]
  15. Duan, T.; Du, Y.; Xing, C.; Wang, H.Y.; Wang, R.F. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front. Immunol. 2022, 13, 812774. [Google Scholar] [CrossRef] [PubMed]
  16. Samuelsson, P.; Hang, L.; Wullt, B.; Irjala, H.; Svanborg, C. Toll-like receptor 4 expression and cytokine responses in the human urinary tract mucosa. Infect. Immun. 2004, 72, 3179–3186. [Google Scholar] [CrossRef]
  17. Liu, M.; Zen, K. Toll-Like Receptors Regulate the Development and Progression of Renal Diseases. Kidney Dis. 2021, 7, 14–23. [Google Scholar] [CrossRef]
  18. Molteni, M.; Gemma, S.; Rossetti, C. The Role of Toll-Like Receptor 4 in Infectious and Noninfectious Inflammation. Mediat. Inflamm. 2016, 2016, 6978936. [Google Scholar] [CrossRef] [PubMed]
  19. Guijarro-Muñoz, I.; Compte, M.; Álvarez-Cienfuegos, A.; Álvarez-Vallina, L.; Sanz, L. Lipopolysaccharide activates Toll-like receptor 4 (TLR4)-mediated NF-κB signaling pathway and proinflammatory response in human pericytes. J. Biol. Chem. 2014, 289, 2457–2468. [Google Scholar] [CrossRef]
  20. Pu, D.; Wang, W. Toll-like receptor 4 agonist, lipopolysaccharide, increases the expression levels of cytokines and chemokines in human peripheral blood mononuclear cells. Exp. Ther. Med. 2014, 8, 1914–1918. [Google Scholar] [CrossRef] [PubMed]
  21. Cheng, L.; Chen, Q.; Zhu, W.; Wu, H.; Wang, Q.; Shi, L.; Zhao, X.; Han, D. Toll-like Receptors 4 and 5 Cooperatively Initiate the Innate Immune Responses to Uropathogenic Escherichia coli Infection in Mouse Epididymal Epithelial Cells1. Biol. Reprod. 2016, 94, 58. [Google Scholar] [CrossRef]
  22. Song, J.; Bishop, B.L.; Li, G.; Grady, R.; Stapleton, A.; Abraham, S.N. TLR4-mediated expulsion of bacteria from infected bladder epithelial cells. Proc. Natl. Acad. Sci. USA 2009, 106, 14966–14971. [Google Scholar] [CrossRef]
  23. Krogfelt, K.A.; Bergmans, H.; Klemm, P. Direct evidence that the FimH protein is the mannose-specific adhesin of Escherichia coli type 1 fimbriae. Infect. Immun. 1990, 58, 1995–1998. [Google Scholar] [CrossRef]
  24. Legros, N.; Ptascheck, S.; Pohlentz, G.; Karch, H.; Dobrindt, U.; Müthing, J. PapG subtype-specific binding characteristics of Escherichia coli towards globo-series glycosphingolipids of human kidney and bladder uroepithelial cells. Glycobiology 2019, 29, 789–802. [Google Scholar] [CrossRef] [PubMed]
  25. Lane, M.C.; Mobley, H.L.T. Role of P-fimbrial-mediated adherence in pyelonephritis and persistence of uropathogenic Escherichia coli (UPEC) in the mammalian kidney. Kidney Int. 2007, 72, 19–25. [Google Scholar] [CrossRef]
  26. Anderson, G.G.; Palermo, J.J.; Schilling, J.D.; Roth, R.; Heuser, J.; Hultgren, S.J. Intracellular bacterial biofilm-like pods in urinary tract infections. Science 2003, 301, 105–107. [Google Scholar] [CrossRef]
  27. Justice, S.S.; Hung, C.; Theriot, J.A.; Fletcher, D.A.; Anderson, G.G.; Footer, M.J.; Hultgren, S.J. Differentiation and developmental pathways of uropathogenic Escherichia coli in urinary tract pathogenesis. Proc. Natl. Acad. Sci. USA 2004, 101, 1333–1338. [Google Scholar] [CrossRef] [PubMed]
  28. Sharma, K.; Dhar, N.; Thacker, V.V.; Simonet, T.M.; Signorino-Gelo, F.; Knott, G.; McKinney, J.D. Dynamic persistence of UPEC intracellular bacterial communities in a human bladder-chip model of urinary tract infection. eLife 2021, 10, e66481. [Google Scholar] [CrossRef]
  29. Behzadi, P.; Urbán, E.; Gajdács, M. Association between Biofilm-Production and Antibiotic Resistance in Uropathogenic Escherichia coli (UPEC): An In Vitro Study. Diseases 2020, 8, 17. [Google Scholar] [CrossRef] [PubMed]
  30. Tewawong, N.; Kowaboot, S.; Pimainog, Y.; Watanagul, N.; Thongmee, T.; Poovorawan, Y. Distribution of phylogenetic groups, adhesin genes, biofilm formation, and antimicrobial resistance of uropathogenic Escherichia coli isolated from hospitalized patients in Thailand. PeerJ 2020, 8, e10453. [Google Scholar] [CrossRef]
  31. Qasemi, A.; Rahimi, F.; Katouli, M. Genetic diversity and virulence characteristics of biofilm-producing uropathogenic Escherichia coli. Int. Microbiol. 2022, 25, 297–307. [Google Scholar] [CrossRef]
  32. Król, J.E.; Wojtowicz, A.J.; Rogers, L.M.; Heuer, H.; Smalla, K.; Krone, S.M.; Top, E.M. Invasion of E. coli biofilms by antibiotic resistance plasmids. Plasmid 2013, 70, 110–119. [Google Scholar] [CrossRef] [PubMed]
  33. Massip, C.; Oswald, E. Siderophore-Microcins in Escherichia coli: Determinants of Digestive Colonization, the First Step Toward Virulence. Front. Cell. Infect. Microbiol. 2020, 10, 381. [Google Scholar] [CrossRef]
  34. Wang, C.; Li, Q.; Lv, J.; Sun, X.; Cao, Y.; Yu, K.; Miao, C.; Zhang, Z.S.; Yao, Z.; Wang, Q. Alpha-hemolysin of uropathogenic Escherichia coli induces GM-CSF-mediated acute kidney injury. Mucosal Immunol. 2020, 13, 22–33. [Google Scholar] [CrossRef] [PubMed]
  35. Carlini, F.; Maroccia, Z.; Fiorentini, C.; Travaglione, S.; Fabbri, A. Effects of the Escherichia coli Bacterial Toxin Cytotoxic Necrotizing Factor 1 on Different Human and Animal Cells: A Systematic Review. Int. J. Mol. Sci. 2021, 22, 12610. [Google Scholar] [CrossRef] [PubMed]
  36. Freire, C.A.; Silva, R.M.; Ruiz, R.C.; Pimenta, D.C.; Bryant, J.A.; Henderson, I.R.; Barbosa, A.S.; Elias, W.P. Secreted Autotransporter Toxin (Sat) Mediates Innate Immune System Evasion. Front. Immunol. 2022, 13, 844878. [Google Scholar] [CrossRef]
  37. Dobrindt, U.; Agerer, F.; Michaelis, K.; Janka, A.; Buchrieser, C.; Samuelson, M.; Svanborg, C.; Gottschalk, G.; Karch, H.; Hacker, J. Analysis of genome plasticity in pathogenic and commensal Escherichia coli isolates by use of DNA arrays. J. Bacteriol. 2003, 185, 1831–1840. [Google Scholar] [CrossRef]
  38. Cusumano, C.K.; Hung, C.S.; Chen, S.L.; Hultgren, S.J. Virulence plasmid harbored by uropathogenic Escherichia coli functions in acute stages of pathogenesis. Infect. Immun. 2010, 78, 1457–1467. [Google Scholar] [CrossRef]
  39. Valiatti, T.B.; Santos, F.F.; Santos, A.C.M.; Nascimento, J.A.S.; Silva, R.M.; Carvalho, E.; Sinigaglia, R.; Gomes, T.A.T. Genetic and Virulence Characteristics of a Hybrid Atypical Enteropathogenic and Uropathogenic Escherichia coli (aEPEC/UPEC) Strain. Front. Cell. Infect. Microbiol. 2020, 10, 492. [Google Scholar] [CrossRef]
  40. Nascimento, J.A.S.; Santos, F.F.; Valiatti, T.B.; Santos-Neto, J.F.; M Santos, A.C.; Cayô, R.; Gales, A.C.; Gomes, T.A.T. Frequency and Diversity of Hybrid Escherichia coli Strains Isolated from Urinary Tract Infections. Microorganisms 2021, 9, 693. [Google Scholar] [CrossRef]
  41. Tanabe, R.H.S.; Dias, R.C.B.; Orsi, H.; de Lira, D.R.P.; Vieira, M.A.; Dos Santos, L.F.; Ferreira, A.M.; Rall, V.L.M.; Mondelli, A.L.; Gomes, T.A.T.; et al. Characterization of Uropathogenic Escherichia coli Reveals Hybrid Isolates of Uropathogenic and Diarrheagenic (UPEC/DEC) E. coli. Microorganisms 2022, 10, 645. [Google Scholar] [CrossRef]
  42. Donnenberg, M.S.; Tzipori, S.; McKee, M.L.; O’Brien, A.D.; Alroy, J.; Kaper, J.B. The role of the eae gene of enterohemorrhagic Escherichia coli in intimate attachment in vitro and in a porcine model. J. Clin. Investig. 1993, 92, 1418–1424. [Google Scholar] [CrossRef]
  43. Morin, N.; Santiago, A.E.; Ernst, R.K.; Guillot, S.J.; Nataro, J.P. Characterization of the AggR regulon in enteroaggregative Escherichia coli. Infect. Immun. 2013, 81, 122–132. [Google Scholar] [CrossRef]
  44. Yousefipour, M.; Rezatofighi, S.E.; Ardakani, M.R. Detection and characterization of hybrid uropathogenic Escherichia coli strains among E. coli isolates causing community-acquired urinary tract infection. J. Med. Microbiol. 2023, 72, 001660. [Google Scholar] [CrossRef]
  45. Clermont, O.; Christenson, J.K.; Denamur, E.; Gordon, D.M. The Clermont Escherichia coli phylo-typing method revisited: Improvement of specificity and detection of new phylo-groups. Environ. Microbiol. Rep. 2013, 5, 58–65. [Google Scholar] [CrossRef] [PubMed]
  46. de Castro Stoppe, N.; Silva, J.S.; Carlos, C.; Sato, M.I.Z.; Saraiva, A.M.; Ottoboni, L.M.M.; Torres, T.T. Worldwide phylogenetic group patterns of Escherichia coli from commensal human and wastewater treatment plant isolates. Front. Microbiol. 2017, 8, 2512. [Google Scholar] [CrossRef] [PubMed]
  47. Chakraborty, A.; Saralaya, V.; Adhikari, P.; Shenoy, S.; Baliga, S.; Hegde, A. Characterization of Escherichia coli Phylogenetic Groups Associated with Extraintestinal Infections in South Indian Population. Ann. Med. Health Sci. Res. 2015, 5, 241–246. [Google Scholar] [CrossRef] [PubMed]
  48. Halaji, M.; Fayyazi, A.; Rajabnia, M.; Zare, D.; Pournajaf, A.; Ranjbar, R. Phylogenetic Group Distribution of Uropathogenic Escherichia coli and Related Antimicrobial Resistance Pattern: A Meta-Analysis and Systematic Review. Front. Cell. Infect. Microbiol. 2022, 12, 790184. [Google Scholar] [CrossRef]
  49. Fratamico, P.M.; DebRoy, C.; Liu, Y.; Needleman, D.S.; Baranzoni, G.M.; Feng, P. Advances in Molecular Serotyping and Subtyping of Escherichia coli. Front. Microbiol. 2016, 7, 644. [Google Scholar] [CrossRef]
  50. Abe, C.M.; Salvador, F.A.; Falsetti, I.N.; Vieira, M.A.M.; Blanco, J.; Blanco, J.E.; Blanco, M.; Machado, A.M.O.; Elias, W.P.; Hernandes, R.T.; et al. Uropathogenic Escherichia coli (UPEC) strains may carry virulence properties of diarrhoeagenic E. coli. FEMS Immunol. Med. Microbiol. 2008, 52, 397–406. [Google Scholar] [CrossRef]
  51. Yamamoto, S. Molecular epidemiology of uropathogenic Escherichia coli. J. Infect. Chemother. 2007, 13, 68–73. [Google Scholar] [CrossRef]
  52. Paniagua-Contreras, G.L.; Monroy-Pérez, E.; Rodríguez-Moctezuma, J.R.; Domínguez-Trejo, P.; Vaca-Paniagua, F.; Vaca, S. Virulence factors, antibiotic resistance phenotypes and O-serogroups of Escherichia coli strains isolated from community-acquired urinary tract infection patients in Mexico. J. Microbiol. Immunol. Infect. 2017, 50, 478–485. [Google Scholar] [CrossRef]
  53. Mohammed, E.J.; Hasan, K.C.; Allami, M. Phylogenetic groups, serogroups and virulence factors of uropathogenic Escherichia coli isolated from patients with urinary tract infection in Baghdad, Iraq. Iran. J. Microbiol. 2022, 14, 445–457. [Google Scholar] [CrossRef]
  54. Prakapaite, R.; Saab, F.; Planciuniene, R.; Petraitis, V.; Walsh, T.J.; Petraitiene, R.; Semoskaite, R.; Baneviciene, R.; Kalediene, L.; Kavaliauskas, P. Molecular Characterization of Uropathogenic Escherichia coli Reveals Emergence of Drug Resistant O15, O22 and O25 Serogroups. Medicina 2019, 55, 733. [Google Scholar] [CrossRef]
  55. Noie Oskouie, A.; Hasani, A.; Ahangarzadeh Rezaee, M.; Soroush Bar Haghi, M.H.; Hasani, A.; Soltani, E. A Relationship between O-Serotype, Antibiotic Susceptibility and Biofilm Formation in Uropathogenic Escherichia coli. Microb. Drug Resist. 2019, 25, 951–958. [Google Scholar] [CrossRef]
  56. Dormanesh, B.; Safarpoor Dehkordi, F.; Hosseini, S.; Momtaz, H.; Mirnejad, R.; Hoseini, M.J.; Yahaghi, E.; Tarhriz, V.; Khodaverdi Darian, E. Virulence factors and o-serogroups profiles of uropathogenic Escherichia coli isolated from Iranian pediatric patients. Iran. Red Crescent Med. J. 2014, 16, e14627. [Google Scholar] [CrossRef]
  57. O’Grady, M.C.; Barry, L.; Corcoran, G.D.; Hooton, C.; Sleator, R.D.; Lucey, B. Empirical treatment of urinary tract infections: How rational are our guidelines? J. Antimicrob. Chemother. 2018, 74, 214–217. [Google Scholar] [CrossRef]
  58. Neupane, S.; Pant, N.D.; Khatiwada, S.; Chaudhary, R.; Banjara, M.R. Correlation between biofilm formation and resistance toward different commonly used antibiotics along with extended spectrum beta lactamase production in uropathogenic Escherichia coli isolated from the patients suspected of urinary tract infections visiting Shree Birendra Hospital, Chhauni, Kathmandu, Nepal. Antimicrob. Resist. Infect. Control 2016, 5, 5. [Google Scholar] [CrossRef]
  59. Eberly, A.R.; Floyd, K.A.; Beebout, C.J.; Colling, S.J.; Fitzgerald, M.J.; Stratton, C.W.; Schmitz, J.E.; Hadjifrangiskou, M. Biofilm Formation by Uropathogenic Escherichia coli Is Favored under Oxygen Conditions That Mimic the Bladder Environment. Int. J. Mol. Sci. 2017, 18, 2077. [Google Scholar] [CrossRef]
  60. Rahdar, M.; Rashki, A.; Miri, H.R.; Ghalehnoo, M.R. Detection of pap, sfa, afa, foc, and fim adhesin-encoding operons in uropathogenic Escherichia coli isolates collected from patients with urinary tract infection. Jundishapur J. Microbiol. 2015, 8, e22647. [Google Scholar] [CrossRef]
  61. Kadry, A.A.; Al-Kashef, N.M.; El-Ganiny, A.M. Distribution of genes encoding adhesins and biofilm formation capacity among Uropathogenic Escherichia coli isolates in relation to the antimicrobial resistance. Afr. Health Sci. 2020, 20, 238. [Google Scholar] [CrossRef]
  62. Schüroff, P.A.; Abe, C.M.; Silva, J.W.; de Paula Coelho, C.; Andrade, F.B.; Hernandes, R.T.; Dobrindt, U.; Gomes, T.A.T.; Elias, W.P. Role of aggregate-forming pilus (AFP) in adherence and colonization of both intestinal and urinary tracts. Virulence 2022, 13, 1423–1433. [Google Scholar] [CrossRef]
  63. Wojciuk, B.; Majewska, K.; Grygorcewicz, B.; Krukowska, Ż.; Kwiatkowska, E.; Ciechanowski, K.; Dołęgowska, B. The role of uropathogenic Escherichia coli adhesive molecules in inflammatory response-comparative study on immunocompetent hosts and kidney recipients. PLoS ONE 2022, 17, e0268243. [Google Scholar] [CrossRef]
  64. Nhu, N.T.K.; Phan, M.D.; Peters, K.M.; Lo, A.W.; Forde, B.M.; Chong, T.M.; Yin, W.F.; Chan, K.G.; Chromek, M.; Brauner, A.; et al. Discovery of new genes involved in curli production by a uropathogenic Escherichia coli strain from the highly virulent O45:K1:H7 lineage. MBio 2018, 9, 1462–1480. [Google Scholar] [CrossRef]
  65. Barnhart, M.M.; Chapman, M.R. Curli Biogenesis and Function. Annu. Rev. Microbiol. 2006, 60, 131. [Google Scholar] [CrossRef]
  66. Das Mitra, S.; Irshad, P.; Anusree, M.; Rekha, I.; Shailaja, S.; Suresh, J.; Aishwarya, G.; Shrestha, S.; Shome, B.R. Whole genome global insight of antibiotic resistance gene repertoire and virulome of high-risk multidrug-resistant Uropathogenic Escherichia coli. Microb. Pathog. 2021, 161, 105256. [Google Scholar] [CrossRef]
  67. Zamani, H.; Salehzadeh, A. Biofilm formation in uropathogenic Escherichia coli: Association with adhesion factor genes. Turkish J. Med. Sci. 2018, 48, 162–167. [Google Scholar] [CrossRef]
  68. Yazdi, M.; Bouzari, M.; Ghaemi, E.A. Detection of fim, pap, sfa and afa Adhesin-Encoding Operons in Escherichia coli Strains Isolated from Urinary Tract Infections. Med. Lab. J. 2018, 12, 10–15. [Google Scholar] [CrossRef]
  69. Kerrn, M.B.; Struve, C.; Blom, J.; Frimodt-Møller, N.; Krogfelt, K.A. Intracellular persistence of Escherichia coli in urinary bladders from mecillinam-treated mice. J. Antimicrob. Chemother. 2005, 55, 383–386. [Google Scholar] [CrossRef] [PubMed]
  70. Mysorekar, I.U.; Hultgren, S.J. Mechanisms of uropathogenic Escherichia coli persistence and eradication from the urinary tract. Proc. Natl. Acad. Sci. USA 2006, 103, 14170–14175. [Google Scholar] [CrossRef]
  71. Sivick, K.E.; Mobley, H.L.T. Waging war against uropathogenic Escherichia coli: Winning back the urinary tract. Infect. Immun. 2010, 78, 568–585. [Google Scholar] [CrossRef]
  72. Schwan, W.R. Regulation of fim genes in uropathogenic Escherichia coli. World J. Clin. Infect. Dis. 2011, 1, 17–25. [Google Scholar] [CrossRef] [PubMed]
  73. Melican, K.; Sandoval, R.M.; Kader, A.; Josefsson, L.; Tanner, G.A.; Molitoris, B.A.; Richter-Dahlfors, A. Uropathogenic Escherichia coli P and Type 1 fimbriae act in synergy in a living host to facilitate renal colonization leading to nephron obstruction. PLoS Pathog. 2011, 7, e1001298. [Google Scholar] [CrossRef]
  74. Blomfield, I.C. The regulation of pap and type 1 fimbriation in Escherichia coli. Adv. Microb. Physiol. 2001, 45, 1–49. [Google Scholar] [CrossRef]
  75. van der Woude, M.W.; Bäumler, A.J. Phase and antigenic variation in bacteria. Clin. Microbiol. Rev. 2004, 17, 581–611. [Google Scholar] [CrossRef] [PubMed]
  76. Totsika, M.; Beatson, S.A.; Holden, N.; Gally, D.L. Regulatory interplay between pap operons in uropathogenic Escherichia coli. Mol. Microbiol. 2008, 67, 996–1011. [Google Scholar] [CrossRef]
  77. Abraham, J.M.; Freitag, C.S.; Clements, J.R.; Eisenstein, B.I. An invertible element of DNA controls phase variation of type 1 fimbriae of Escherichia coli. Proc. Natl. Acad. Sci. USA 1985, 82, 5724–5727. [Google Scholar] [CrossRef]
  78. Bessaiah, H.; Anamalé, C.; Sung, J.; Dozois, C.M. What Flips the Switch? Signals and Stress Regulating Extraintestinal Pathogenic Escherichia coli Type 1 Fimbriae (Pili). Microorganisms 2022, 10, 5. [Google Scholar] [CrossRef]
  79. Dalebroux, Z.D.; Svensson, S.L.; Gaynor, E.C.; Swanson, M.S. ppGpp conjures bacterial virulence. Microbiol. Mol. Biol. Rev. 2010, 74, 171–199. [Google Scholar] [CrossRef]
  80. Yang, J.; Anderson, B.W.; Turdiev, A.; Turdiev, H.; Stevenson, D.M.; Amador-Noguez, D.; Lee, V.T.; Wang, J.D. The nucleotide pGpp acts as a third alarmone in Bacillus, with functions distinct from those of (p) ppGpp. Nat. Commun. 2020, 11, 5388. [Google Scholar] [CrossRef] [PubMed]
  81. Karczewska, M.; Strzelecki, P.; Bogucka, K.; Potrykus, K.; Szalewska-Pałasz, A.; Nowicki, D. Increased Levels of (p)ppGpp Correlate with Virulence and Biofilm Formation, but Not with Growth, in Strains of Uropathogenic Escherichia coli. Int. J. Mol. Sci. 2023, 24, 3315. [Google Scholar] [CrossRef]
  82. Qin, X.; Hu, F.; Wu, S.; Ye, X.; Zhu, D.; Zhang, Y.; Wang, M. Comparison of Adhesin Genes and Antimicrobial Susceptibilities between Uropathogenic and Intestinal Commensal Escherichia coli Strains. PLoS ONE 2013, 8, e61169. [Google Scholar] [CrossRef]
  83. Källenius, G.; Möllby, R.; Svenson, S.B.; Helin, I.; Hultberg, H.; Cedergren, B.; Winberg, J. Occurrence of P-fimbriated Escherichia coli in urinary tract infections. Lancet 1981, 2, 1369–1372. [Google Scholar] [CrossRef] [PubMed]
  84. Kudinha, T.; Kong, F. Distribution of papG alleles among uropathogenic Escherichia coli from reproductive age women. J. Biomed. Sci. 2022, 29, 1–12. [Google Scholar] [CrossRef] [PubMed]
  85. Johnson, J.R.; Russo, T.A.; Brown, J.J.; Stapleton, A. papG alleles of Escherichia coli strains causing first-episode or recurrent acute cystitis in adult women. J. Infect. Dis. 1998, 177, 97–101. [Google Scholar] [CrossRef]
  86. Nou, X.; Skinner, B.; Braaten, B.; Blyn, L.; Hirsch, D.; Low, D. Regulation of pyelonephritis-associated pili phase-variation in Escherichia coli: Binding of the PapI and the Lrp regulatory proteins is controlled by DNA methylation. Mol. Microbiol. 1993, 7, 545–553. [Google Scholar] [CrossRef]
  87. Hernday, A.D.; Braaten, B.A.; Low, D.A. The mechanism by which DNA adenine methylase and PapI activate the pap epigenetic switch. Mol. Cell 2003, 12, 947–957. [Google Scholar] [CrossRef]
  88. Forsman, K.; Göransson, M.; Uhlin, B.E. Autoregulation and multiple DNA interactions by a transcriptional regulatory protein in E. coli pili biogenesis. EMBO J. 1989, 8, 1271–1277. [Google Scholar] [CrossRef] [PubMed]
  89. Behzadi, P. Uropathogenic Escherichia coli and Fimbrial Adhesins Virulome. In Urinary Tract Infection; Jarzembowski, T., Daca, A., Dębska-Ślizień, M.A., Eds.; IntechOpen: Rijeka, Croatia, 2017. [Google Scholar]
  90. Balsalobre, C.; Morschhäuser, J.; Hacker, J.; Uhlin, B.E. Transcriptional analysis of the SFA and PAP determinants of uropathogenic Escherichia coli strains. Adv. Exp. Med. Biol. 2000, 485, 119–122. [Google Scholar]
  91. Sabitha, B.; Vimal, K.; Geetha, R. Adhesins of Uropathogenic Escherichia coli (UPEC). IP Int. J. Med. Microbiol. Trop. Dis. 2016, 2, 10–18. [Google Scholar]
  92. Korhonen, T.K.; Väisänen-Rhen, V.; Rhen, M.; Pere, A.; Parkkinen, J.; Finne, J. Escherichia coli fimbriae recognizing sialyl galactosides. J. Bacteriol. 1984, 159, 762–766. [Google Scholar] [CrossRef]
  93. Prasadarao, N.V.; Wass, C.A.; Kim, K.S. Identification and characterization of S fimbria-binding sialoglycoproteins on brain microvascular endothelial cells. Infect. Immun. 1997, 65, 2852–2860. [Google Scholar] [CrossRef]
  94. Tajbakhsh, E.; Ahmadi, P.; Abedpour-Dehkordi, E.; Arbab-Soleimani, N.; Khamesipour, F. Biofilm formation, antimicrobial susceptibility, serogroups and virulence genes of uropathogenic E. coli isolated from clinical samples in Iran. Antimicrob. Resist. Infect. Control 2016, 5, 11. [Google Scholar] [CrossRef]
  95. Naves, P.; del Prado, G.; Huelves, L.; Gracia, M.; Ruiz, V.; Blanco, J.; Dahbi, G.; Blanco, M.; del Carmen Ponte, M.; Soriano, F. Correlation between virulence factors and in vitro biofilm formation by Escherichia coli strains. Microb. Pathog. 2008, 45, 86–91. [Google Scholar] [CrossRef]
  96. Servin, A.L. Pathogenesis of Afa/Dr diffusely adhering Escherichia coli. Clin. Microbiol. Rev. 2005, 18, 264–292. [Google Scholar] [CrossRef] [PubMed]
  97. Archambaud, M.; Courcoux, P.; Labigne-Roussel, A. Detection by molecular hybridization of pap, afa, and sfa adherence systems in Escherichia coli strains associated with urinary and enteral infections. Ann. Inst. Pasteur. Microbiol. 1988, 139, 575–588. [Google Scholar] [CrossRef] [PubMed]
  98. Foxman, B.; Zhang, L.; Tallman, P.; Palin, K.; Rode, C.; Bloch, C.; Gillespie, B.; Marrs, C.F. Virulence characteristics of Escherichia coli causing first urinary tract infection predict risk of second infection. J. Infect. Dis. 1995, 172, 1536–1541. [Google Scholar] [CrossRef]
  99. Alvarez-Fraga, L.; Phan, M.-D.; Goh, K.G.K.; Nhu, N.T.K.; Hancock, S.J.; Allsopp, L.P.; Peters, K.M.; Forde, B.M.; Roberts, L.W.; Sullivan, M.J.; et al. Differential Afa/Dr Fimbriae Expression in the Multidrug-Resistant Escherichia coli ST131 Clone. MBio 2022, 13, e0351921. [Google Scholar] [CrossRef]
  100. Nowicki, B.; Selvarangan, R.; Nowicki, S. Family of Escherichia coli Dr adhesins: Decay-accelerating factor receptor recognition and invasiveness. J. Infect. Dis. 2001, 183 (Suppl. 1), S24–S27. [Google Scholar] [CrossRef]
  101. Korotkova, N.; Yarova-Yarovaya, Y.; Tchesnokova, V.; Yazvenko, N.; Carl, M.A.; Stapleton, A.E.; Moseley, S.L. Escherichia coli DraE adhesin-associated bacterial internalization by epithelial cells is promoted independently by decay-accelerating factor and carcinoembryonic antigen-related cell adhesion molecule binding and does not require the DraD invasin. Infect. Immun. 2008, 76, 3869–3880. [Google Scholar] [CrossRef] [PubMed]
  102. Das, M.; Hart-Van Tassell, A.; Urvil, P.T.; Lea, S.; Pettigrew, D.; Anderson, K.L.; Samet, A.; Kur, J.; Matthews, S.; Nowicki, S.; et al. Hydrophilic domain II of Escherichia coli Dr fimbriae facilitates cell invasion. Infect. Immun. 2005, 73, 6119–6126. [Google Scholar] [CrossRef]
  103. van der Woude, M.W.; Low, D.A. Leucine-responsive regulatory protein and deoxyadenosine methylase control the phase variation and expression of the sfa and daa pili operons in Escherichia coli. Mol. Microbiol. 1994, 11, 605–618. [Google Scholar] [CrossRef]
  104. Benz, R. RTX-Toxins. Toxins 2020, 12, 359. [Google Scholar] [CrossRef]
  105. Guo, S.; Vance, T.D.R.; Stevens, C.A.; Voets, I.; Davies, P.L. RTX Adhesins are Key Bacterial Surface Megaproteins in the Formation of Biofilms. Trends Microbiol. 2019, 27, 453–467. [Google Scholar] [CrossRef]
  106. Vigil, P.D.; Wiles, T.J.; Engstrom, M.D.; Prasov, L.; Mulvey, M.A.; Mobley, H.L.T. The repeat-in-toxin family member TosA mediates adherence of uropathogenic Escherichia coli and survival during bacteremia. Infect. Immun. 2012, 80, 493–505. [Google Scholar] [CrossRef] [PubMed]
  107. Luterbach, C.L.; Forsyth, V.S.; Engstrom, M.D.; Mobley, H.L.T. TosR-Mediated Regulation of Adhesins and Biofilm Formation in Uropathogenic Escherichia coli. mSphere 2018, 3, e00222-18. [Google Scholar] [CrossRef]
  108. Miller, A.L.; Bessho, S.; Grando, K.; Tükel, Ç. Microbiome or Infections: Amyloid-Containing Biofilms as a Trigger for Complex Human Diseases. Front. Immunol. 2021, 12, 638867. [Google Scholar] [CrossRef]
  109. Bhoite, S.; van Gerven, N.; Chapman, M.R.; Remaut, H. Curli Biogenesis: Bacterial Amyloid Assembly by the Type VIII Secretion Pathway. EcoSal Plus 2019, 8, 10–128. [Google Scholar] [CrossRef] [PubMed]
  110. Evans, M.L.; Chapman, M.R. Curli biogenesis: Order out of disorder. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 1551–1558. [Google Scholar] [CrossRef]
  111. Homma, M.; Kojima, S. Roles of the second messenger c-di-GMP in bacteria: Focusing on the topics of flagellar regulation and Vibrio spp. Genes Cells 2022, 27, 157–172. [Google Scholar] [CrossRef] [PubMed]
  112. Ross, P.; Mayer, R.; Weinhouse, H.; Amikam, D.; Huggirat, Y.; Benziman, M.; de Vroom, E.; Fidder, A.; de Paus, P.; Sliedregt, L.A. The cyclic diguanylic acid regulatory system of cellulose synthesis in Acetobacter xylinum. Chemical synthesis and biological activity of cyclic nucleotide dimer, trimer, and phosphothioate derivatives. J. Biol. Chem. 1990, 265, 18933–18943. [Google Scholar] [CrossRef]
  113. Cepas, V.; Ballén, V.; Gabasa, Y.; Ramírez, M.; López, Y.; Soto, S.M. Transposon Insertion in the purL Gene Induces Biofilm Depletion in Escherichia coli ATCC 25922. Pathogens 2020, 9, 774. [Google Scholar] [CrossRef] [PubMed]
  114. Morar, M.; Hoskins, A.A.; Stubbe, J.; Ealick, S.E. Formylglycinamide ribonucleotide amidotransferase from Thermotoga maritima: Structural insights into complex formation. Biochemistry 2008, 47, 7816–7830. [Google Scholar] [CrossRef] [PubMed]
  115. Martín-Rodríguez, A.J.; Rhen, M.; Melican, K.; Richter-Dahlfors, A. Nitrate Metabolism Modulates Biosynthesis of Biofilm Components in Uropathogenic Escherichia coli and Acts as a Fitness Factor During Experimental Urinary Tract Infection. Front. Microbiol. 2020, 11, 26. [Google Scholar] [CrossRef] [PubMed]
  116. Holden, E.R.; Yasir, M.; Turner, A.K.; Wain, J.; Charles, I.G.; Webber, M.A. Massively parallel transposon mutagenesis identifies temporally essential genes for biofilm formation in Escherichia coli. Microb. Genom. 2021, 7, 000673. [Google Scholar] [CrossRef] [PubMed]
  117. Javed, S.; Mirani, Z.A.; Pirzada, Z.A. Phylogenetic Group B2 Expressed Significant Biofilm Formation among Drug Resistant Uropathogenic Escherichia coli. Libyan J. Med. 2021, 16. [Google Scholar] [CrossRef]
  118. Ochoa, S.A.; Cruz-Córdova, A.; Luna-Pineda, V.M.; Reyes-Grajeda, J.P.; Cázares-Domínguez, V.; Escalona, G.; Sepúlveda-González, M.E.; López-Montiel, F.; Arellano-Galindo, J.; López-Martínez, B.; et al. Multidrug- and Extensively Drug-Resistant Uropathogenic Escherichia coli Clinical Strains: Phylogenetic Groups Widely Associated with Integrons Maintain High Genetic Diversity. Front. Microbiol. 2016, 7, 2042. [Google Scholar] [CrossRef]
  119. Whelan, S.; O’Grady, M.C.; Corcoran, D.; Finn, K.; Lucey, B. Uropathogenic Escherichia coli Biofilm-Forming Capabilities are not Predictable from Clinical Details or from Colonial Morphology. Diseases 2020, 8, 11. [Google Scholar] [CrossRef]
  120. Whelan, S.; O’Grady, M.C.; Corcoran, G.D.; Finn, K.; Lucey, B. Effect of Sub-Inhibitory Concentrations of Nitrofurantoin, Ciprofloxacin, and Trimethoprim on In Vitro Biofilm Formation in Uropathogenic Escherichia coli (UPEC). Med. Sci. 2022, 11, 1. [Google Scholar] [CrossRef]
  121. Yasmeen, K.; Chunchanur, S.K.; Nadagir, S.D.; Halesh, L.H.; Chandrasekhar, M.R. Virulence factors, serotypes and antimicrobial suspectibility pattern of Escherichia coli in urinary tract infections. Al Ameen J. Med. Sci. 2009, 2, 47–51. [Google Scholar]
  122. Karam, M.R.A.; Habibi, M.; Bouzari, S. Relationships between Virulence Factors and Antimicrobial Resistance among Escherichia coli Isolated from Urinary Tract Infections and Commensal Isolates in Tehran, Iran. Osong Public Health Res. Perspect. 2018, 9, 217–224. [Google Scholar] [CrossRef]
  123. Shah, C.; Baral, R.; Bartaula, B.; Shrestha, L.B. Virulence factors of uropathogenic Escherichia coli (UPEC) and correlation with antimicrobial resistance. BMC Microbiol. 2019, 19, 204. [Google Scholar] [CrossRef]
  124. Radera, S.; Srivastava, S.; Agarwal, J. Virulence Genotyping and Multidrug Resistance Pattern of Escherichia coli Isolated From Community-Acquired and Hospital-Acquired Urinary Tract Infections. Cureus 2022, 14, e29404. [Google Scholar] [CrossRef] [PubMed]
  125. Stanley, P.; Koronakis, V.; Hughes, C. Acylation of Escherichia coli hemolysin: A unique protein lipidation mechanism underlying toxin function. Microbiol. Mol. Biol. Rev. 1998, 62, 309–333. [Google Scholar] [CrossRef] [PubMed]
  126. Felmlee, T.; Pellett, S.; Welch, R.A. Nucleotide sequence of an Escherichia coli chromosomal hemolysin. J. Bacteriol. 1985, 163, 94–105. [Google Scholar] [CrossRef]
  127. Wiles, T.J.; Dhakal, B.K.; Eto, D.S.; Mulvey, M.A. Inactivation of host Akt/protein kinase B signaling by bacterial pore-forming toxins. Mol. Biol. Cell 2008, 19, 1427–1438. [Google Scholar] [CrossRef]
  128. Zhang, Z.; Wang, M.; Zhang, Y.; Zhang, Y.; Bartkuhn, M.; Markmann, M.; Hossain, H.; Chakraborty, T.; Hake, S.B.; Jia, Z.; et al. Uropathogenic Escherichia coli Virulence Factor α-Hemolysin Reduces Histone Acetylation to Inhibit Expression of Proinflammatory Cytokine Genes. J. Infect. Dis. 2021, 223, 1040–1051. [Google Scholar] [CrossRef]
  129. Doye, A.; Mettouchi, A.; Bossis, G.; Clément, R.; Buisson-Touati, C.; Flatau, G.; Gagnoux, L.; Piechaczyk, M.; Boquet, P.; Lemichez, E. CNF1 Exploits the Ubiquitin-Proteasome Machinery to Restrict Rho GTPase Activation for Bacterial Host Cell Invasion. Cell 2002, 111, 553–564. [Google Scholar] [CrossRef] [PubMed]
  130. Lerm, M.; Schmidt, G.; Aktories, K. Bacterial protein toxins targeting rho GTPases. FEMS Microbiol. Lett. 2000, 188, 1–6. [Google Scholar] [CrossRef]
  131. Villalonga, P.; Ridley, A.J. Rho GTPases and cell cycle control. Growth Factors 2006, 24, 159–164. [Google Scholar] [CrossRef]
  132. Mao, Y.; Finnemann, S.C. Regulation of phagocytosis by Rho GTPases. Small GTPases 2015, 6, 89–99. [Google Scholar] [CrossRef]
  133. Oswald, E.; Sugai, M.; Labigne, A.; Wu, H.C.; Fiorentini, C.; Boquet, P.; O’Brien, A.D. Cytotoxic necrotizing factor type 2 produced by virulent Escherichia coli modifies the small GTP-binding proteins Rho involved in assembly of actin stress fibers. Proc. Natl. Acad. Sci. USA 1994, 91, 3814–3818. [Google Scholar] [CrossRef]
  134. Sugai, M.; Hatazaki, K.; Mogami, A.; Ohta, H.; Pérès, S.Y.; Hérault, F.; Horiguchi, Y.; Masuda, M.; Ueno, Y.; Komatsuzawa, H.; et al. Cytotoxic necrotizing factor type 2 produced by pathogenic Escherichia coli deamidates a Gln residue in the conserved G-3 domain of the rho family and preferentially inhibits the GTPase activity of RhoA and Rac1. Infect. Immun. 1999, 67, 6550–6557. [Google Scholar] [CrossRef]
  135. Oswald, E.; De Rycke, J. A single protein of 110 kDa is associated with the multinucleating and necrotizing activity coded by the Vir plasmid of Escherichia coli. FEMS Microbiol. Lett. 1990, 56, 279–284. [Google Scholar]
  136. Blum, G.; Falbo, V.; Caprioli, A.; Hacker, J. Gene clusters encoding the cytotoxic necrotizing factor type 1, Prs-fimbriae and α-hemolysin form the pathogenicity island II of the uropathogenic Escherichia coli strain J96. FEMS Microbiol. Lett. 1995, 126, 189–195. [Google Scholar] [CrossRef]
  137. McNichol, B.A.; Rasmussen, S.B.; Carvalho, H.M.; Meysick, K.C.; O’Brien, A.D. Two domains of cytotoxic necrotizing factor type 1 bind the cellular receptor, laminin receptor precursor protein. Infect. Immun. 2007, 75, 5095–5104. [Google Scholar] [CrossRef] [PubMed]
  138. Chung, J.W.; Hong, S.J.; Kim, K.J.; Goti, D.; Stins, M.F.; Shin, S.; Dawson, V.L.; Dawson, T.M.; Kim, K.S. 37-kDa laminin receptor precursor modulates cytotoxic necrotizing factor 1-mediated RhoA activation and bacterial uptake. J. Biol. Chem. 2003, 278, 16857–16862. [Google Scholar] [CrossRef] [PubMed]
  139. Yamamoto, S.; Tsukamoto, T.; Terai, A.; Kurazono, H.; Takeda, Y.; Yoshida, O. Distribution of virulence factors in Escherichia coli isolated from urine of cystitis patients. Microbiol. Immunol. 1995, 39, 401–404. [Google Scholar] [CrossRef]
  140. Rippere-Lampe, K.E.; O’Brien, A.D.; Conran, R.; Lockman, H.A. Mutation of the gene encoding cytotoxic necrotizing factor type 1 (cnf(1)) attenuates the virulence of uropathogenic Escherichia coli. Infect. Immun. 2001, 69, 3954–3964. [Google Scholar] [CrossRef] [PubMed]
  141. Elliott, S.J.; Srinivas, S.; Albert, M.J.; Alam, K.; Robins-Browne, R.M.; Gunzburg, S.T.; Mee, B.J.; Chang, B.J. Characterization of the roles of hemolysin and other toxins in enteropathy caused by alpha-hemolytic Escherichia coli linked to human diarrhea. Infect. Immun. 1998, 66, 2040–2051. [Google Scholar] [CrossRef] [PubMed]
  142. Landraud, L.; Gauthier, M.; Fosse, T.; Boquet, P. Frequency of Escherichia coli strains producing the cytotoxic necrotizing factor (CNF1) in nosocomial urinary tract infections. Lett. Appl. Microbiol. 2000, 30, 213–216. [Google Scholar] [CrossRef]
  143. Wang, M.-H.; Kim, K.S. Cytotoxic necrotizing factor 1 contributes to Escherichia coli meningitis. Toxins 2013, 5, 2270–2280. [Google Scholar] [CrossRef] [PubMed]
  144. Chang, A.C.; Krishnan, S.; Prasadarao, N.V. The effects of cytotoxic necrotizing factor 1 expression in the uptake of Escherichia coli K1 by macrophages and the onset of meningitis in newborn mice. Virulence 2016, 7, 806–818. [Google Scholar] [CrossRef]
  145. Pokharel, P.; Habouria, H.; Bessaiah, H.; Dozois, C.M. Serine Protease Autotransporters of the Enterobacteriaceae (SPATEs): Out and About and Chopping It Up. Microorganisms 2019, 7, 594. [Google Scholar] [CrossRef]
  146. Guyer, D.M.; Henderson, I.R.; Nataro, J.P.; Mobley, H.L.T. Identification of Sat, an autotransporter toxin produced by uropathogenic Escherichia coli. Mol. Microbiol. 2000, 38, 53–66. [Google Scholar] [CrossRef] [PubMed]
  147. Guyer, D.M.; Radulovic, S.; Jones, F.-E.; Mobley, H.L.T. Sat, the secreted autotransporter toxin of uropathogenic Escherichia coli, is a vacuolating cytotoxin for bladder and kidney epithelial cells. Infect. Immun. 2002, 70, 4539–4546. [Google Scholar] [CrossRef]
  148. Cover, T.L. The vacuolating cytotoxin of Helicobacter pylori. Mol. Microbiol. 1996, 20, 241–246. [Google Scholar] [CrossRef]
  149. Balsalobre, C.; Morschhäuser, J.; Jass, J.; Hacker, J.; Uhlin, B.E. Transcriptional analysis of the sfa determinant revealing mmRNA processing events in the biogenesis of S fimbriae in pathogenic Escherichia coli. J. Bacteriol. 2003, 185, 620–629. [Google Scholar] [CrossRef]
  150. Saha, R.; Saha, N.; Donofrio, R.S.; Bestervelt, L.L. Microbial siderophores: A mini review. J. Basic Microbiol. 2013, 53, 303–317. [Google Scholar] [CrossRef] [PubMed]
  151. Andreini, C.; Bertini, I.; Cavallaro, G.; Holliday, G.L.; Thornton, J.M. Metal ions in biological catalysis: From enzyme databases to general principles. J. Biol. Inorg. Chem. 2008, 13, 1205–1218. [Google Scholar] [CrossRef]
  152. Nairz, M.; Dichtl, S.; Schroll, A.; Haschka, D.; Tymoszuk, P.; Theurl, I.; Weiss, G. Iron and innate antimicrobial immunity—Depriving the pathogen, defending the host. J. Trace Elem. Med. Biol. 2018, 48, 118–133. [Google Scholar] [CrossRef]
  153. Hood, M.I.; Skaar, E.P. Nutritional immunity: Transition metals at the pathogen–host interface. Nat. Rev. Microbiol. 2012, 10, 525–537. [Google Scholar] [CrossRef] [PubMed]
  154. Gao, M.; Zhao, T.; Zhang, C.; Li, P.; Wang, J.; Han, J.; Zhang, N.; Pang, B.; Liu, S. Ferritinophagy-mediated iron competition in RUTIs: Tug-of-war between UPEC and host. Biomed. Pharmacother. 2023, 163, 114859. [Google Scholar] [CrossRef] [PubMed]
  155. Henderson, J.P.; Crowley, J.R.; Pinkner, J.S.; Walker, J.N.; Tsukayama, P.; Stamm, W.E.; Hooton, T.M.; Hultgren, S.J. Quantitative metabolomics reveals an epigenetic blueprint for iron acquisition in uropathogenic Escherichia coli. PLoS Pathog. 2009, 5, e1000305. [Google Scholar] [CrossRef]
  156. Robinson, A.E.; Heffernan, J.R.; Henderson, J.P. The iron hand of uropathogenic Escherichia coli: The role of transition metal control in virulence. Future Microbiol. 2018, 13, 745–756. [Google Scholar] [CrossRef] [PubMed]
  157. Goetz, D.H.; Holmes, M.A.; Borregaard, N.; Bluhm, M.E.; Raymond, K.N.; Strong, R.K. The neutrophil lipocalin NGAL is a bacteriostatic agent that interferes with siderophore-mediated iron acquisition. Mol. Cell 2002, 10, 1033–1043. [Google Scholar] [CrossRef]
  158. Rezatofighi, S.E.; Mirzarazi, M.; Salehi, M. Virulence genes and phylogenetic groups of uropathogenic Escherichia coli isolates from patients with urinary tract infection and uninfected control subjects: A case-control study. BMC Infect. Dis. 2021, 21, 361. [Google Scholar] [CrossRef]
  159. Torres, A.G.; Payne, S.M. Haem iron-transport system in enterohaemorrhagic Escherichia coli O157:H7. Mol. Microbiol. 1997, 23, 825–833. [Google Scholar] [CrossRef]
  160. Hagan, E.C.; Mobley, H.L.T. Haem acquisition is facilitated by a novel receptor Hma and required by uropathogenic Escherichia coli for kidney infection. Mol. Microbiol. 2009, 71, 79–91. [Google Scholar] [CrossRef]
  161. Roos, V.; Ulett, G.C.; Schembri, M.A.; Klemm, P. The asymptomatic bacteriuria Escherichia coli strain 83972 outcompetes uropathogenic E. coli strains in human urine. Infect. Immun. 2006, 74, 615–624. [Google Scholar] [CrossRef]
  162. Bush, K.; Bradford, P.A. β-Lactams and β-Lactamase Inhibitors: An Overview. Cold Spring Harb. Perspect. Med. 2016, 6, a025247. [Google Scholar] [CrossRef]
  163. Halaji, M.; Shahidi, S.; Atapour, A.; Ataei, B.; Feizi, A.; Havaei, S.A. Characterization of Extended-Spectrum β-Lactamase-Producing Uropathogenic Escherichia coli Among Iranian Kidney Transplant Patients. Infect. Drug Resist. 2020, 13, 1429–1437. [Google Scholar] [CrossRef]
  164. Lange, F.; Pfennigwerth, N.; Höfken, L.-M.; Gatermann, S.G.; Kaase, M. Characterization of mutations in Escherichia coli PBP2 leading to increased carbapenem MICs. J. Antimicrob. Chemother. 2019, 74, 571–576. [Google Scholar] [CrossRef]
  165. Chetri, S.; Bhowmik, D.; Paul, D.; Pandey, P.; Chanda, D.D.; Chakravarty, A.; Bora, D.; Bhattacharjee, A. AcrAB-TolC efflux pump system plays a role in carbapenem non-susceptibility in Escherichia coli. BMC Microbiol. 2019, 19, 210. [Google Scholar] [CrossRef]
  166. Rossolini, G.M.; D’Andrea, M.M.; Mugnaioli, C. The Spread of CTX-M-Type Extended-Spectrum β-Lactamases. Clin. Microbiol. Infect. 2008, 14, 33–41. [Google Scholar] [CrossRef]
  167. Shrestha, R.; Khanal, S.; Poudel, P.; Khadayat, K.; Ghaju, S.; Bhandari, A.; Lekhak, S.; Pant, N.D.; Sharma, M.; Marasini, B.P. Extended spectrum β-lactamase producing uropathogenic Escherichia coli and the correlation of biofilm with antibiotics resistance in Nepal. Ann. Clin. Microbiol. Antimicrob. 2019, 18, 42. [Google Scholar] [CrossRef] [PubMed]
  168. Bush, K.; Bradford, P.A. Epidemiology of β-Lactamase-Producing Pathogens. Clin. Microbiol. Rev. 2020, 3. [Google Scholar] [CrossRef] [PubMed]
  169. Aryal, S.C.; Upreti, M.K.; Sah, A.K.; Ansari, M.; Nepal, K.; Dhungel, B.; Adhikari, N.; Lekhak, B.; Rijal, K.R. Plasmid-Mediated AmpC β-Lactamase CITM and DHAM Genes Among Gram-Negative Clinical Isolates. Infect. Drug Resist. 2020, 13, 4249–4261. [Google Scholar] [CrossRef] [PubMed]
  170. Tracz, D.M.; Boyd, D.A.; Hizon, R.; Bryce, E.; McGeer, A.; Ofner-Agostini, M.; Simor, A.E.; Paton, S.; Mulvey, M.R.; Program, C.N.I.S. ampC gene expression in promoter mutants of cefoxitin-resistant Escherichia coli clinical isolates. FEMS Microbiol. Lett. 2007, 270, 265–271. [Google Scholar] [CrossRef] [PubMed]
  171. Sheu, C.-C.; Chang, Y.-T.; Lin, S.-Y.; Chen, Y.-H.; Hsueh, P.-R. Infections Caused by Carbapenem-Resistant Enterobacteriaceae: An Update on Therapeutic Options. Front. Microbiol. 2019, 10, 80. [Google Scholar] [CrossRef]
  172. Zangane Matin, F.; Rezatofighi, S.E.; Roayaei Ardakani, M.; Akhoond, M.R.; Mahmoodi, F. Virulence characterization and clonal analysis of uropathogenic Escherichia coli metallo-beta-lactamase-producing isolates. Ann. Clin. Microbiol. Antimicrob. 2021, 20, 50. [Google Scholar] [CrossRef]
  173. Ranjitkar, S.; Reck, F.; Ke, X.; Zhu, Q.; McEnroe, G.; Lopez, S.L.; Dean, C.R. Identification of Mutations in the mrdA Gene Encoding PBP2 That Reduce Carbapenem and Diazabicyclooctane Susceptibility of Escherichia coli Clinical Isolates with Mutations in ftsI (PBP3) and Which Carry bla(NDM-1). mSphere 2019, 4, e00074-19. [Google Scholar] [CrossRef]
  174. van Duin, D.; Bonomo, R.A. Ceftazidime/Avibactam and Ceftolozane/Tazobactam: Second-generation β-Lactam/β-Lactamase Inhibitor Combinations. Clin. Infect. Dis. 2016, 63, 234–241. [Google Scholar] [CrossRef] [PubMed]
  175. Yahav, D.; Giske, C.G.; Grāmatniece, A.; Abodakpi, H.; Tam, V.H.; Leibovici, L. New β-Lactam-β-Lactamase Inhibitor Combinations. Clin. Microbiol. Rev. 2020, 34. [Google Scholar] [CrossRef] [PubMed]
  176. Chen, M.; Zhang, M.; Huang, P.; Lin, Q.; Sun, C.; Zeng, H.; Deng, Y. Novel β-lactam/β-lactamase inhibitors versus alternative antibiotics for the treatment of complicated intra-abdominal infection and complicated urinary tract infection: A meta-analysis of randomized controlled trials. Expert Rev. Anti-Infect. Ther. 2018, 16, 111–120. [Google Scholar] [CrossRef]
  177. Zhang, Y.; Kashikar, A.; Brown, C.A.; Denys, G.; Bush, K. Unusual Escherichia coli PBP 3 Insertion Sequence Identified from a Collection of Carbapenem-Resistant Enterobacteriaceae Tested In Vitro with a Combination of Ceftazidime-, Ceftaroline-, or Aztreonam-Avibactam. Antimicrob. Agents Chemother. 2017, 61, 10–128. [Google Scholar] [CrossRef]
  178. Li, J.; Xu, Q.; Ogurek, S.; Li, Z.; Wang, P.; Xie, Q.; Sheng, Z.; Wang, M. Efflux Pump AcrAB Confers Decreased Susceptibility to Piperacillin-Tazobactam and Ceftolozane-Tazobactam in Tigecycline-Non-Susceptible Klebsiella pneumoniae. Infect. Drug Resist. 2020, 13, 4309–4319. [Google Scholar] [CrossRef] [PubMed]
  179. Tutone, M.; Bjerklund Johansen, T.E.; Cai, T.; Mushtaq, S.; Livermore, D.M. SUsceptibility and Resistance to Fosfomycin and other antimicrobial agents among pathogens causing lower urinary tract infections: Findings of the SURF study. Int. J. Antimicrob. Agents 2022, 59, 106574. [Google Scholar] [CrossRef] [PubMed]
  180. McOsker, C.C.; Fitzpatrick, P.M. Nitrofurantoin: Mechanism of action and implications for resistance development in common uropathogens. J. Antimicrob. Chemother. 1994, 33, 23–30. [Google Scholar] [CrossRef]
  181. Whiteway, J.; Koziarz, P.; Veall, J.; Sandhu, N.; Kumar, P.; Hoecher, B.; Lambert, I.B. Oxygen-insensitive nitroreductases: Analysis of the roles of nfsA and nfsB in development of resistance to 5-nitrofuran derivatives in Escherichia coli. J. Bacteriol. 1998, 180, 5529–5539. [Google Scholar] [CrossRef]
  182. Wan, Y.; Mills, E.; Leung, R.C.Y.; Vieira, A.; Zhi, X.; Croucher, N.J.; Woodford, N.; Jauneikaite, E.; Ellington, M.J.; Sriskandan, S. Alterations in chromosomal genes nfsA, nfsB, and ribE are associated with nitrofurantoin resistance in Escherichia coli from the United Kingdom. Microb. Genom. 2021, 7, 702. [Google Scholar] [CrossRef]
  183. Vervoort, J.; Xavier, B.B.; Stewardson, A.; Coenen, S.; Godycki-Cwirko, M.; Adriaenssens, N.; Kowalczyk, A.; Lammens, C.; Harbarth, S.; Goossens, H.; et al. An In Vitro Deletion in ribE Encoding Lumazine Synthase Contributes to Nitrofurantoin Resistance in Escherichia coli. Antimicrob. Agents Chemother. 2014, 58, 7225. [Google Scholar] [CrossRef]
  184. Li, J.; Zhang, H.; Ning, J.; Sajid, A.; Cheng, G.; Yuan, Z.; Hao, H. The nature and epidemiology of OqxAB, a multidrug efflux pump. Antimicrob. Resist. Infect. Control. 2019, 8, 44. [Google Scholar] [CrossRef] [PubMed]
  185. Ho, P.L.; Ng, K.Y.; Lo, W.U.; Law, P.Y.; Lai, E.L.Y.; Wang, Y.; Chow, K.H. Plasmid-Mediated OqxAB Is an Important Mechanism for Nitrofurantoin Resistance in Escherichia coli. Antimicrob. Agents Chemother. 2016, 60, 537. [Google Scholar] [CrossRef]
  186. Khamari, B.; Kumar, P.; Pradeep, B.E. Resistance to nitrofurantoin is an indicator of extensive drug-resistant (XDR) Enterobacteriaceae. J. Med. Microbiol. 2021, 70, 001347. [Google Scholar] [CrossRef] [PubMed]
  187. Edowik, Y.; Caspari, T.; Williams, H.M. The amino acid changes t55a, a273p and r277c in the beta-lactamase CTX-M-14 render E. coli resistant to the antibiotic nitrofurantoin, a first-line treatment of urinary tract infections. Microorganisms 2020, 8, 1983. [Google Scholar] [CrossRef]
  188. Dzib-Baak, H.E.; Uc-Cachón, A.H.; de Jesús Dzul-Beh, A.; Rosado-Manzano, R.F.; Gracida-Osorno, C.; Molina-Salinas, G.M. Efficacy of Fosfomycin against Planktonic and Biofilm-Associated MDR Uropathogenic Escherichia coli Clinical Isolates. Trop. Med. Infect. Dis. 2022, 7, 235. [Google Scholar] [CrossRef]
  189. Falagas, M.E.; Athanasaki, F.; Voulgaris, G.L.; Triarides, N.A.; Vardakas, K.Z. Resistance to fosfomycin: Mechanisms, Frequency and Clinical Consequences. Int. J. Antimicrob. Agents 2019, 53, 22–28. [Google Scholar] [CrossRef] [PubMed]
  190. Ríos, E.; del Carmen López Diaz, M.; Culebras, E.; Rodríguez-Avial, I.; Rodríguez-Avial, C. Resistance to fosfomycin is increasing and is significantly associated with extended-spectrum β-lactamase-production in urinary isolates of Escherichia coli. Med. Microbiol. Immunol. 2022, 211, 269–272. [Google Scholar] [CrossRef]
  191. Silver, L.L. Fosfomycin: Mechanism and Resistance. Cold Spring Harb. Perspect. Med. 2017, 7, a025262. [Google Scholar] [CrossRef]
  192. Fu, Z.; Ma, Y.; Chen, C.; Guo, Y.; Hu, F.; Liu, Y.; Xu, X.; Wang, M. Prevalence of Fosfomycin Resistance and Mutations in murA, glpT, and uhpT in Methicillin-Resistant Staphylococcus aureus Strains Isolated from Blood and Cerebrospinal Fluid Samples. Front. Microbiol. 2015, 6, 1544. [Google Scholar] [CrossRef] [PubMed]
  193. Ambudkar, S.V.; Larson, T.J.; Maloney, P.C. Reconstitution of sugar phosphate transport systems of Escherichia coli. J. Biol. Chem. 1986, 261, 9083–9086. [Google Scholar] [CrossRef] [PubMed]
  194. Ballestero-Téllez, M.; Docobo-Pérez, F.; Portillo-Calderón, I.; Rodríguez-Martínez, J.M.; Racero, L.; Ramos-Guelfo, M.S.; Blázquez, J.; Rodríguez-Baño, J.; Pascual, A. Molecular insights into fosfomycin resistance in Escherichia coli. J. Antimicrob. Chemother. 2017, 72, 1303–1309. [Google Scholar] [CrossRef] [PubMed]
  195. Galindo-Méndez, M.; Navarrete-Salazar, H.; Baltazar-Jiménez, F.; Muñoz-de la Paz, E.; Sánchez-Mawcinitt, M.F.; Gómez-Pardo, A.; Garza-González, E.; Ponce-de-León-Garduño, L.A.; Franco-Cendejas, R.; Morfín-Otero, R.; et al. Emergence of Fosfomycin Resistance by Plasmid-Mediated fos Genes in Uropathogenic ESBL-Producing E. coli Isolates in Mexico. Antibiotics 2022, 11, 1383. [Google Scholar] [CrossRef]
  196. Poirel, L.; Vuillemin, X.; Kieffer, N.; Mueller, L.; Descombes, M.C.; Nordmann, P. Identification of FosA8, a Plasmid-Encoded Fosfomycin Resistance Determinant from Escherichia coli, and Its Origin in Leclercia adecarboxylata. Antimicrob. Agents Chemother. 2019, 63, e01403-19. [Google Scholar] [CrossRef]
  197. Kieffer, N.; Poirel, L.; Descombes, M.C.; Nordmann, P. Characterization of FosL1, a Plasmid-Encoded Fosfomycin Resistance Protein Identified in Escherichia coli. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef]
  198. López-Montesinos, I.; Horcajada, J.P. Oral and intravenous fosfomycin in complicated urinary tract infections. Rev. Esp. Quimioter. 2019, 32 (Suppl. 1), 37–44. [Google Scholar] [PubMed]
  199. Bergan, T.; Thorsteinsson, S.B.; Albini, E. Pharmacokinetic profile of fosfomycin trometamol. Chemotherapy 1993, 39, 297–301. [Google Scholar] [CrossRef]
  200. Marino, A.; Stracquadanio, S.; Ceccarelli, M.; Zagami, A.; Nunnari, G.; Cacopardo, B. Oral fosfomycin formulation for acute bacterial prostatitis; a new role for an old molecule: A case report and brief literature review. World Acad. Sci. J. 2022, 4, 1–6. [Google Scholar] [CrossRef]
  201. Gardiner, B.J.; Mahony, A.A.; Ellis, A.G.; Lawrentschuk, N.; Bolton, D.M.; Zeglinski, P.T.; Frauman, A.G.; Grayson, M.L. Is Fosfomycin a Potential Treatment Alternative for Multidrug-Resistant Gram-Negative Prostatitis? Clin. Infect. Dis. 2013, 58, e101–e105. [Google Scholar] [CrossRef]
  202. Hashemian, S.M.R.; Farhadi, Z.; Farhadi, T. Fosfomycin: The characteristics, activity, and use in critical care. Ther. Clin. Risk Manag. 2019, 15, 525–530. [Google Scholar] [CrossRef]
  203. Marino, A.; Stracquadanio, S.; Campanella, E.; Munafò, A.; Gussio, M.; Ceccarelli, M.; Bernardini, R.; Nunnari, G.; Cacopardo, B. Intravenous Fosfomycin: A Potential Good Partner for Cefiderocol. Clinical Experience and Considerations. Antibiotics 2023, 12, 49. [Google Scholar] [CrossRef]
  204. Masters, P.A.; O’Bryan, T.A.; Zurlo, J.; Miller, D.Q.; Joshi, N. Trimethoprim-Sulfamethoxazole Revisited. Arch. Intern. Med. 2003, 163, 402–410. [Google Scholar] [CrossRef]
  205. Mahmood, M.S.; Ibrahim, A.H. Investigation of the Gene Expression of Class I Integron and Its Relationship to Antibiotic Resistance in Isolates of Escherichia coli Causing Urinary Tract Diseases. HIV Nurs. 2023, 23, 705–710. [Google Scholar]
  206. Stephens, C.; Arismendi, T.; Wright, M.; Hartman, A.; Gonzalez, A.; Gill, M.; Pandori, M.; Hess, D. F Plasmids Are the Major Carriers of Antibiotic Resistance Genes in Human-Associated Commensal Escherichia coli. mSphere 2020, 5. [Google Scholar] [CrossRef] [PubMed]
  207. Sánchez-Osuna, M.; Cortés, P.; Llagostera, M.; Barbé, J.; Erill, I. Exploration into the origins and mobilization of di-hydrofolate reductase genes and the emergence of clinical resistance to trimethoprim. Microb. Genom. 2020, 6, mgen000440. [Google Scholar] [CrossRef] [PubMed]
  208. Al-Assil, B.; Mahfoud, M.; Hamzeh, A.R. First report on class 1 integrons and Trimethoprim-resistance genes from dfrA group in uropathogenic E. coli (UPEC) from the Aleppo area in Syria. Mob. Genet. Elements 2013, 3, e25204. [Google Scholar] [CrossRef] [PubMed]
  209. Li, D.; Reid, C.J.; Kudinha, T.; Jarocki, V.M.; Djordjevic, S.P. Genomic analysis of trimethoprim-resistant extraintestinal pathogenic Escherichia coli and recurrent urinary tract infections. Microb. Genom. 2020, 6, mgen000475. [Google Scholar] [CrossRef]
  210. Rahman, M.M.; Hossain, M.M.K.; Rubaya, R.; Halder, J.; Karim, M.E.; Bhuiya, A.A.; Khatun, A.; Alam, J. Association of Antibiotic Resistance Traits in Uropathogenic Escherichia coli (UPEC) Isolates. Can. J. Infect. Dis. Med. Microbiol. 2022, 2022, 4251486. [Google Scholar] [CrossRef]
  211. Zeng, Q.; Xiao, S.; Gu, F.; He, W.; Xie, Q.; Yu, F.; Han, L. Antimicrobial Resistance and Molecular Epidemiology of Uropathogenic Escherichia coli Isolated From Female Patients in Shanghai, China. Front. Cell. Infect. Microbiol. 2021, 11, 653983. [Google Scholar] [CrossRef]
  212. Mulder, M.; Verbon, A.; Lous, J.; Goessens, W.; Stricker, B.H. Use of other antimicrobial drugs is associated with trimethoprim resistance in patients with urinary tract infections caused by E. coli. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 2283. [Google Scholar] [CrossRef]
  213. Song, Y.; Zhou, Z.; Gu, J.; Yang, J.; Deng, J. Reducing the Periplasmic Glutathione Content Makes Escherichia coli Resistant to Trimethoprim and Other Antimicrobial Drugs. Microbiol. Spectr. 2021, 9, e00743-21. [Google Scholar] [CrossRef] [PubMed]
  214. Shi, H.; Li, T.; Xu, J.; Yu, J.; Yang, S.; Zhang, X.E.; Tao, S.; Gu, J.; Deng, J.Y. MgrB Inactivation Confers Trimethoprim Resistance in Escherichia coli. Front. Microbiol. 2021, 12, 682205. [Google Scholar] [CrossRef]
  215. Chao, Y.-S.; Farrah, K. Fluoroquinolones for the Treatment of Urinary Tract Infection 2; Canadian Agency for Drugs and Technologies in Health: Ottawa, ON, Canada, 2019. [Google Scholar]
  216. Blondeau, J.M. Fluoroquinolones: Mechanism of action, classification, and development of resistance. Surv. Ophthalmol. 2004, 49, S73–S78. [Google Scholar] [CrossRef] [PubMed]
  217. Esmaeel, N.E.; Gerges, M.A.; Hosny, T.A.; Ali, A.R.; Gebriel, M.G. Detection of Chromosomal and Plasmid-Mediated Quinolone Resistance Among Escherichia coli Isolated from Urinary Tract Infection Cases; Zagazig University Hospitals, Egypt. Infect. Drug Resist. 2020, 13, 413–421. [Google Scholar] [CrossRef] [PubMed]
  218. Huguet, A.; Pensec, J.; Soumet, C. Resistance in Escherichia coli: Variable contribution of efflux pumps with respect to different fluoroquinolones. J. Appl. Microbiol. 2013, 114, 1294–1299. [Google Scholar] [CrossRef]
  219. Sato, T.; Yokota, S.-I.; Uchida, I.; Okubo, T.; Usui, M.; Kusumoto, M.; Akiba, M.; Fujii, N.; Tamura, Y. Fluoroquinolone resistance mechanisms in an Escherichia coli isolate, HUE1, without quinolone resistance-determining region mutations. Front. Microbiol. 2013, 4, 125. [Google Scholar] [CrossRef] [PubMed]
  220. Salah, F.D.; Soubeiga, S.T.; Ouattara, A.K.; Sadji, A.Y.; Metuor-Dabire, A.; Obiri-Yeboah, D.; Banla-Kere, A.; Karou, S.; Simpore, J. Distribution of quinolone resistance gene (qnr) in ESBL-producing Escherichia coli and Klebsiella spp. in Lomé, Togo. Antimicrob. Resist. Infect. Control 2019, 8, 104. [Google Scholar] [CrossRef] [PubMed]
  221. Shetty, S.; Deekshit, V.K.; Jazeela, K.; Vittal, R.; Rohit, A.; Chakraborty, A.; Karunasagar, I. Plasmid-mediated fluoroquinolone resistance associated with extra-intestinal Escherichia coli isolates from hospital samples. Indian J. Med. Res. 2019, 149, 192. [Google Scholar] [CrossRef]
  222. Kuo, P.Y.; Lo, Y.T.; Chiou, Y.J.; Chen, C.A.; Hidrosollo, J.H.; Thuy, T.T.D.; Zhang, Y.Z.; Wang, M.C.; Lin, T.P.; Lin, W.H.; et al. Plasmid-mediated quinolone resistance determinants in fluoroquinolone-nonsusceptible Escherichia coli isolated from patients with urinary tract infections in a university hospital, 2009–2010 and 2020. J. Glob. Antimicrob. Resist. 2022, 30, 241–248. [Google Scholar] [CrossRef]
  223. Phan, M.D.; Peters, K.M.; Fraga, L.A.; Wallis, S.C.; Hancock, S.J.; Nhu, N.T.K.; Forde, B.M.; Bauer, M.J.; Paterson, D.L.; Beatson, S.A.; et al. Plasmid-Mediated Ciprofloxacin Resistance Imparts a Selective Advantage on Escherichia coli ST131. Antimicrob. Agents Chemother. 2022, 66, e02146-21. [Google Scholar] [CrossRef]
  224. Machuca, J.; Ortiz, M.; Recacha, E.; Díaz-De-Alba, P.; Docobo-Perez, F.; Rodríguez-Martínez, J.-M.; Pascual, Á. Impact of AAC(6′)-Ib-cr in combination with chromosomal-mediated mechanisms on clinical quinolone resistance in Escherichia coli. J. Antimicrob. Chemother. 2016, 71, 3066–3071. [Google Scholar] [CrossRef] [PubMed]
  225. Zurfluh, K.; Treier, A.; Schmitt, K.; Stephan, R. Mobile fosfomycin resistance genes in Enterobacteriaceae—An increasing threat. Microbiologyopen 2020, 9, e1135. [Google Scholar] [CrossRef] [PubMed]
  226. Lee, J.C.; Oh, J.Y.; Cho, J.W.; Park, J.C.; Kim, J.M.; Seol, S.Y.; Cho, D.T. The prevalence of trimethoprim-resistance-conferring dihydrofolate reductase genes in urinary isolates of Escherichia coli in Korea. J. Antimicrob. Chemother. 2001, 47, 599–604. [Google Scholar] [CrossRef]
  227. Brolund, A.; Sundqvist, M.; Kahlmeter, G.; Grape, M. Molecular characterisation of trimethoprim resistance in Escherichia coli and Klebsiella pneumoniae during a two year intervention on trimethoprim use. PLoS ONE 2010, 5, e9233. [Google Scholar] [CrossRef] [PubMed]
  228. Rijavec, M.; Starcic Erjavec, M.; Ambrozic Avgustin, J.; Reissbrodt, R.; Fruth, A.; Krizan-Hergouth, V.; Zgur-Bertok, D. High prevalence of multidrug resistance and random distribution of mobile genetic elements among uropathogenic Escherichia coli (UPEC) of the four major phylogenetic groups. Curr. Microbiol. 2006, 53, 158–162. [Google Scholar] [CrossRef]
  229. Kuznetsova, M.V.; Gizatullina, J.S. Epidemiological characteristics of uropatogenic isolates of Escherichia coli in hospitals. Klin. Lab. Diagn. 2021, 66, 248–256. [Google Scholar] [CrossRef]
  230. Carattoli, A.; Bertini, A.; Villa, L.; Falbo, V.; Hopkins, K.L.; Threlfall, E.J. Identification of plasmids by PCR-based replicon typing. J. Microbiol. Methods 2005, 63, 219–228. [Google Scholar] [CrossRef]
  231. Bousquet, A.; Henquet, S.; Compain, F.; Genel, N.; Arlet, G.; Decré, D. Partition locus-based classification of selected plasmids in Klebsiella pneumoniae, Escherichia coli and Salmonella enterica spp.: An additional tool. J. Microbiol. Methods 2015, 110, 85–91. [Google Scholar] [CrossRef]
  232. Garcillán-Barcia, M.P.; Francia, M.V.; de La Cruz, F. The diversity of conjugative relaxases and its application in plasmid classification. FEMS Microbiol. Rev. 2009, 33, 657–687. [Google Scholar] [CrossRef]
  233. García-Fernández, A.; Carattoli, A. Plasmid double locus sequence typing for IncHI2 plasmids, a subtyping scheme for the characterization of IncHI2 plasmids carrying extended-spectrum β-lactamase and quinolone resistance genes. J. Antimicrob. Chemother. 2010, 65, 1155–1161. [Google Scholar] [CrossRef]
  234. Johnson, T.J.; Nolan, L.K. Plasmid replicon typing. Methods Mol. Biol. 2009, 551, 27–35. [Google Scholar] [PubMed]
  235. Rozwandowicz, M.; Brouwer, M.S.M.; Fischer, J.; Wagenaar, J.A.; Gonzalez-Zorn, B.; Guerra, B.; Mevius, D.J.; Hordijk, J. Plasmids carrying antimicrobial resistance genes in Enterobacteriaceae. J. Antimicrob. Chemother. 2018, 73, 1121–1137. [Google Scholar] [CrossRef]
  236. Johnson, T.J.; Wannemuehler, Y.M.; Johnson, S.J.; Logue, C.M.; White, D.G.; Doetkott, C.; Nolan, L.K. Plasmid replicon typing of commensal and pathogenic Escherichia coli isolates. Appl. Environ. Microbiol. 2007, 73, 1976–1983. [Google Scholar] [CrossRef]
  237. Smith, H.; Bossers, A.; Harders, F.; Wu, G.; Woodford, N.; Schwarz, S.; Guerra, B.; Rodríguez, I.; van Essen-Zandbergen, A.; Brouwer, M.; et al. Characterization of epidemic IncI1-Iγ plasmids harboring ambler class A and C genes in Escherichia coli and Salmonella enterica from animals and humans. Antimicrob. Agents Chemother. 2015, 59, 5357–5365. [Google Scholar] [CrossRef]
  238. Tijet, N.; Faccone, D.; Rapoport, M.; Seah, C.; Pasterán, F.; Ceriana, P.; Albornoz, E.; Corso, A.; Petroni, A.; Melano, R.G. Molecular characteristics of mcr-1-carrying plasmids and new mcr-1 variant recovered from polyclonal clinical Escherichia coli from Argentina and Canada. PLoS ONE 2017, 12, e0180347. [Google Scholar] [CrossRef]
  239. Yang, Q.-E.; Sun, J.; Li, L.; Deng, H.; Liu, B.-T.; Fang, L.-X.; Liao, X.-P.; Liu, Y.-H. IncF plasmid diversity in multi-drug resistant Escherichia coli strains from animals in China. Front. Microbiol. 2015, 6, 964. [Google Scholar] [CrossRef] [PubMed]
  240. Tewari, R.; Ganaie, F.; Venugopal, N.; Mitra, S.; Shome, R.; Shome, B.R. Occurrence and characterization of genetic determinants of β-lactam-resistance in Escherichia coli clinical isolates. Infect. Genet. Evol. 2022, 100, 105257. [Google Scholar] [CrossRef]
  241. Soleimani, N.; Derakhshan, S.; Memariani, M. Plasmid Profile Analysis of Aminoglycoside-Resistant Escherichia coli Isolated From Urinary Tract Infections. Int. J. Enteric Pathog. 2016, 4, e33806. [Google Scholar] [CrossRef]
  242. Bean, D.C.; Livermore, D.M.; Hall, L.M.C. Plasmids imparting sulfonamide resistance in Escherichia coli: Implications for persistence. Antimicrob. Agents Chemother. 2009, 53, 1088–1093. [Google Scholar] [CrossRef] [PubMed]
  243. Sun, J.; Chen, C.; Cui, C.-Y.; Zhang, Y.; Liu, X.; Cui, Z.-H.; Ma, X.-Y.; Feng, Y.; Fang, L.-X.; Lian, X.-L.; et al. Plasmid-encoded tet(X) genes that confer high-level tigecycline resistance in Escherichia coli. Nat. Microbiol. 2019, 4, 1457–1464. [Google Scholar] [CrossRef]
  244. Basu, S.; Mukherjee, M. Incidence and risk of co-transmission of plasmid-mediated quinolone resistance and extended-spectrum β-lactamase genes in fluoroquinolone-resistant uropathogenic Escherichia coli: A first study from Kolkata, India. J. Glob. Antimicrob. Resist. 2018, 14, 217–223. [Google Scholar] [CrossRef] [PubMed]
  245. Król, J.E.; Nguyen, H.D.; Rogers, L.M.; Beyenal, H.; Krone, S.M.; Top, E.M. Increased transfer of a multidrug resistance plasmid in Escherichia coli biofilms at the air-liquid interface. Appl. Environ. Microbiol. 2011, 77, 5079–5088. [Google Scholar] [CrossRef]
  246. Kuznetsova, M.V.; Maslennikova, I.L.; Pospelova, J.S.; Žgur Bertok, D.; Starčič Erjavec, M. Differences in recipient ability of uropathogenic Escherichia coli strains in relation with their pathogenic potential. Infect. Genet. Evol. 2022, 97, 105160. [Google Scholar] [CrossRef] [PubMed]
  247. Li, W.; Ma, J.; Sun, X.; Liu, M.; Wang, H. Antimicrobial Resistance and Molecular Characterization of Gene Cassettes from Class 1 Integrons in Escherichia coli Strains. Microb. Drug Resist. 2022, 28, 413. [Google Scholar] [CrossRef] [PubMed]
  248. Oliveira-Pinto, C.; Diamantino, C.; Oliveira, P.L.; Reis, M.P.; Costa, P.S.; Paiva, M.C.; Nardi, R.M.D.; Magalhães, P.P.; Chartone-Souza, E.; Nascimento, A.M.A. Occurrence and characterization of class 1 integrons in Escherichia coli from healthy individuals and those with urinary infection. J. Med. Microbiol. 2017, 66, 577–583. [Google Scholar] [CrossRef]
  249. Javed, S.; Mirani, Z.A.; Pirzada, Z.A. Study of class 1 integrons and plasmid profile among multiple drug resistant uropathogenic Escherichia coli. Pak. J. Pharm. Sci. 2020, 33, 2643–2649. [Google Scholar] [CrossRef]
  250. González-Villalobos, E.; Ribas-Aparicio, R.M.; Belmont-Monroy, L.; Aparicio-Ozores, G.; Manjarrez-Hernández, H.Á.; Gavilanes-Parra, S.; Balcázar, J.L.; Molina-López, J. Identification and characterization of class 1 integrons among multidrug-resistant uropathogenic Escherichia coli strains in Mexico. Microb. Pathog. 2022, 162, 105348. [Google Scholar] [CrossRef]
  251. Fallah, F.; Karimi, A.; Goudarzi, M.; Shiva, F.; Navidinia, M.; Hadipour Jahromi, M.; Sajadi Nia, R.S. Determination of integron frequency by a polymerase chain reaction-restriction fragment length polymorphism method in multidrug-resistant Escherichia coli, which causes urinary tract infections. Microb. Drug Resist. 2012, 18, 546–549. [Google Scholar] [CrossRef]
  252. de los Santos, E.; Laviña, M.; Poey, M.E. Strict relationship between class 1 integrons and resistance to sulfamethoxazole in Escherichia coli. Microb. Pathog. 2021, 161, 105206. [Google Scholar] [CrossRef]
  253. Rozwadowski, M.; Gawel, D. Molecular Factors and Mechanisms Driving Multidrug Resistance in Uropathogenic Escherichia coli—An Update. Genes 2022, 13, 1397. [Google Scholar] [CrossRef]
  254. Pérez-Etayo, L.; Berzosa, M.; González, D.; Vitas, A.I. Prevalence of Integrons and Insertion Sequences in ESBL-Producing E. coli Isolated from Different Sources in Navarra, Spain. Int. J. Environ. Res. Public Health 2018, 15, 2308. [Google Scholar] [CrossRef] [PubMed]
  255. Carvalho, R.; Aburjaile, F.; Canario, M.; Nascimento, A.M.A.; Chartone-Souza, E.; de Jesus, L.; Zamyatnin, A.A.; Brenig, B.; Barh, D.; Ghosh, P.; et al. Genomic Characterization of Multidrug-Resistant Escherichia coli BH100 Sub-strains. Front. Microbiol. 2021, 11, 549254. [Google Scholar] [CrossRef] [PubMed]
  256. Schink, A.K.; Kadlec, K.; Schwarz, S. Detection of qnr genes among Escherichia coli isolates of animal origin and complete sequence of the conjugative qnrB19-carrying plasmid pQNR2078. J. Antimicrob. Chemother. 2012, 67, 1099–1102. [Google Scholar] [CrossRef]
  257. Pan, Y.; Zhang, T.; Yu, L.; Zong, Z.; Zhao, S.; Li, R.; Wang, Q.; Yuan, L.; Hu, G.; He, D. IS 1294 Reorganizes Plasmids in a Multidrug-Resistant Escherichia coli Strain. Microbiol. Spectr. 2021, 9, e00503-21. [Google Scholar] [CrossRef] [PubMed]
  258. Brumbaugh, A.R.; Smith, S.N.; Mobley, H.L.T. Immunization with the yersiniabactin receptor, FyuA, protects against pyelonephritis in a murine model of urinary tract infection. Infect. Immun. 2013, 81, 3309–3316. [Google Scholar] [CrossRef]
  259. Mike, L.A.; Smith, S.N.; Sumner, C.A.; Eaton, K.A.; Mobley, H.L.T. Siderophore vaccine conjugates protect against Uropathogenic Escherichia coli urinary tract infection. Proc. Natl. Acad. Sci. USA 2016, 113, 13468–13473. [Google Scholar] [CrossRef]
  260. Mobley, H.L.T.; Alteri, C.J. Development of a Vaccine against Escherichia coli Urinary Tract Infections. Pathogens 2015, 5, 1. [Google Scholar] [CrossRef]
  261. Forsyth, V.S.; Himpsl, S.D.; Smith, S.N.; Sarkissian, C.A.; Mike, L.A.; Stocki, J.A.; Sintsova, A.; Alteri, C.J.; Mobley, H.L.T. Optimization of an experimental vaccine to prevent Escherichia coli urinary tract infection. MBio 2020, 11, e00555-20. [Google Scholar] [CrossRef]
  262. Wu, J.; Bao, C.; Lee Reinhardt, R.; Abraham, S.N. Local induction of bladder Th1 responses to combat urinary tract infections. Proc. Natl. Acad. Sci. USA 2021, 118, e2026461118. [Google Scholar] [CrossRef]
  263. Lorenzo-Gómez, M.-F.; Foley, S.; Nickel, J.C.; García-Cenador, M.-B.; Padilla-Fernández, B.-Y.; González-Casado, I.; Martínez-Huélamo, M.; Yang, B.; Blick, C.; Ferreira, F.; et al. Sublingual MV140 for Prevention of Recurrent Urinary Tract Infections. NEJM Evid. 2022, 1, EVIDoa2100018. [Google Scholar] [CrossRef]
  264. Kelly, S.H.; Votaw, N.L.; Cossette, B.J.; Wu, Y.; Shetty, S.; Shores, L.S.; Issah, L.A.; Collier, J.H. A sublingual nanofiber vaccine to prevent urinary tract infections. Sci. Adv. 2022, 8, eabq4120. [Google Scholar] [CrossRef]
  265. Butler, D.; Ambite, I.; Wan, M.L.Y.; Tran, T.H.; Wullt, B.; Svanborg, C. Immunomodulation Therapy Offers New Molecular Strategies to Treat UTI. Nat. Rev. Urol. 2022, 19, 419–437. [Google Scholar] [CrossRef] [PubMed]
  266. Puthia, M.; Ambite, I.; Cafaro, C.; Butler, D.; Huang, Y.; Lutay, N.; Rydström, G.; Gullstrand, B.; Swaminathan, B.; Nadeem, A.; et al. IRF7 inhibition prevents destructive innate immunity-A target for nonantibiotic therapy of bacterial infections. Sci. Transl. Med. 2016, 8, 336ra59. [Google Scholar] [CrossRef]
  267. Ambite, I.; Puthia, M.; Nagy, K.; Cafaro, C.; Nadeem, A.; Butler, D.S.C.; Rydström, G.; Filenko, N.A.; Wullt, B.; Miethke, T.; et al. Molecular Basis of Acute Cystitis Reveals Susceptibility Genes and Immunotherapeutic Targets. PLoS Pathog. 2016, 12, e1005848. [Google Scholar] [CrossRef]
  268. de Jonge, P.A.; Nobrega, F.L.; Brouns, S.J.J.; Dutilh, B.E. Molecular and Evolutionary Determinants of Bacteriophage Host Range. Trends Microbiol. 2019, 27, 51–63. [Google Scholar] [CrossRef] [PubMed]
  269. Zulk, J.J.; Clark, J.R.; Ottinger, S.; Ballard, M.B.; Mejia, M.E.; Mercado-Evans, V.; Heckmann, E.R.; Sanchez, B.C.; Trautner, B.W.; Maresso, A.W.; et al. Phage Resistance Accompanies Reduced Fitness of Uropathogenic Escherichia coli in the Urinary Environment. mSphere 2022, 7, e0034522. [Google Scholar] [CrossRef]
  270. Burmeister, A.R.; Fortier, A.; Roush, C.; Lessing, A.J.; Bender, R.G.; Barahman, R.; Grant, R.; Chan, B.K.; Turner, P.E. Pleiotropy complicates a trade-off between phage resistance and antibiotic resistance. Proc. Natl. Acad. Sci. USA 2020, 117, 11207–11216. [Google Scholar] [CrossRef]
  271. Pagnout, C.; Sohm, B.; Razafitianamaharavo, A.; Caillet, C.; Offroy, M.; Leduc, M.; Gendre, H.; Jomini, S.; Beaussart, A.; Bauda, P.; et al. Pleiotropic effects of rfa-gene mutations on Escherichia coli envelope properties. Sci. Rep. 2019, 9, 9696. [Google Scholar] [CrossRef]
  272. Liu, S.; Lu, H.; Zhang, S.; Shi, Y.; Chen, Q. Phages against Pathogenic Bacterial Biofilms and Biofilm-Based Infections: A Review. Pharmaceutics 2022, 14, 427. [Google Scholar] [CrossRef] [PubMed]
  273. Liao, K.S.; Lehman, S.M.; Tweardy, D.J.; Donlan, R.M.; Trautner, B.W. Bacteriophages are synergistic with bacterial interference for the prevention of Pseudomonas aeruginosa biofilm formation on urinary catheters. J. Appl. Microbiol. 2012, 113, 1530–1539. [Google Scholar] [CrossRef]
  274. Sillankorva, S.; Oliveira, D.; Moura, A.; Henriques, M.; Faustino, A.; Nicolau, A.; Azeredo, J. Efficacy of a broad host range lytic bacteriophage against E. coli adhered to urothelium. Curr. Microbiol. 2011, 62, 1128–1132. [Google Scholar] [CrossRef]
  275. Valério, N.; Oliveira, C.; Jesus, V.; Branco, T.; Pereira, C.; Moreirinha, C.; Almeida, A. Effects of single and combined use of bacteriophages and antibiotics to inactivate Escherichia coli. Virus Res. 2017, 240, 8–17. [Google Scholar] [CrossRef] [PubMed]
  276. Bhargava, K.; Nath, G.; Dhameja, N.; Kumar, R.; Aseri, G.K.; Jain, N. Bacteriophage therapy for Escherichia coli-induced urinary tract infection in rats. Future Microbiol. 2023, 18, 323–334. [Google Scholar] [CrossRef]
  277. Mijbel Ali, B.; Gatea Kaabi, S.A.; Al-Bayati, M.A.; Musafer, H.K. A Novel Phage Cocktail Therapy of the Urinary Tract Infection in a Mouse Model. Arch. Razi Inst. 2021, 76, 1229–1236. [Google Scholar] [CrossRef] [PubMed]
  278. Le, T.; Nang, S.C.; Zhao, J.; Yu, H.H.; Li, J.; Gill, J.J.; Liu, M.; Aslam, S. Therapeutic Potential of Intravenous Phage as Standalone Therapy for Recurrent Drug-Resistant Urinary Tract Infections. Antimicrob. Agents Chemother. 2023, 67, e00037-23. [Google Scholar] [CrossRef]
  279. Hitchcock, N.M.; Devequi Gomes Nunes, D.; Shiach, J.; Valeria Saraiva Hodel, K.; Dantas Viana Barbosa, J.; Alencar Pereira Rodrigues, L.; Coler, B.S.; Botelho Pereira Soares, M.; Badaró, R. Current Clinical Landscape and Global Potential of Bacteriophage Therapy. Viruses 2023, 15, 1020. [Google Scholar] [CrossRef]
  280. Loubet, P.; Ranfaing, J.; Dinh, A.; Dunyach-Remy, C.; Bernard, L.; Bruyère, F.; Lavigne, J.P.; Sotto, A. Alternative Therapeutic Options to Antibiotics for the Treatment of Urinary Tract Infections. Front. Microbiol. 2020, 11, 1509. [Google Scholar] [CrossRef]
  281. Greene, S.E.; Pinkner, J.S.; Chorell, E.; Dodson, K.W.; Shaffer, C.L.; Conover, M.S.; Livny, J.; Hadjifrangiskou, M.; Almqvist, F.; Hultgren, S.J. Pilicide ec240 disrupts virulence circuits in uropathogenic Escherichia coli. MBio 2014, 5. [Google Scholar] [CrossRef]
  282. Aberg, V.; Almqvist, F. Pilicides-small molecules targeting bacterial virulence. Org. Biomol. Chem. 2007, 5, 1827–1834. [Google Scholar] [CrossRef]
  283. Cegelski, L.; Pinkner, J.S.; Hammer, N.D.; Cusumano, C.K.; Hung, C.S.; Chorell, E.; Aberg, V.; Walker, J.N.; Seed, P.C.; Almqvist, F.; et al. Small-molecule inhibitors target Escherichia coli amyloid biogenesis and biofilm formation. Nat. Chem. Biol. 2009, 5, 913–919. [Google Scholar] [CrossRef] [PubMed]
  284. Rasmussen, T.B.; Givskov, M. Quorum-sensing inhibitors as anti-pathogenic drugs. Int. J. Med. Microbiol. 2006, 296, 149–161. [Google Scholar] [CrossRef]
  285. Baloyi, I.T.; Cosa, S.; Combrinck, S.; Leonard, C.M.; Viljoen, A.M. Anti-quorum sensing and antimicrobial activities of South African medicinal plants against uropathogens. S. Afr. J. Bot. 2019, 122, 484–491. [Google Scholar] [CrossRef]
  286. Jeevanandam, J.; Chan, Y.S.; Danquah, M.K. Nano-formulations of drugs: Recent developments, impact and challenges. Biochimie 2016, 128–129, 99–112. [Google Scholar] [CrossRef]
  287. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; del Pilar Rodriguez-Torres, M.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef]
  288. Kesisoglou, F.; Panmai, S.; Wu, Y. Nanosizing--oral formulation development and biopharmaceutical evaluation. Adv. Drug Deliv. Rev. 2007, 59, 631–644. [Google Scholar] [CrossRef] [PubMed]
  289. Ghosh, S.; Ghosh, S.; Sil, P.C. Role of nanostructures in improvising oral medicine. Toxicol. Rep. 2019, 6, 358–368. [Google Scholar] [CrossRef]
  290. Wu, Z.-L.; Zhao, J.; Xu, R. Recent Advances in Oral Nano-Antibiotics for Bacterial Infection Therapy. Int. J. Nanomed. 2020, 15, 9587–9610. [Google Scholar] [CrossRef]
  291. Khare, T.; Mahalunkar, S.; Shriram, V.; Gosavi, S.; Kumar, V. Embelin-loaded chitosan gold nanoparticles interact synergistically with ciprofloxacin by inhibiting efflux pumps in multidrug-resistant Pseudomonas aeruginosa and Escherichia coli. Environ. Res. 2021, 199, 111321. [Google Scholar] [CrossRef]
  292. Mukherjee, R.; Patra, M.; Dutta, D.; Banik, M.; Basu, T. Tetracycline-loaded calcium phosphate nanoparticle (Tet-CPNP): Rejuvenation of an obsolete antibiotic to further action. Biochim. Biophys. Acta 2016, 1860, 1929–1941. [Google Scholar] [CrossRef] [PubMed]
  293. Makabenta, J.M.V.; Nabawy, A.; Li, C.-H.; Schmidt-Malan, S.; Patel, R.; Rotello, V.M. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat. Rev. Microbiol. 2021, 19, 23–36. [Google Scholar] [CrossRef] [PubMed]
  294. Qindeel, M.; Barani, M.; Rahdar, A.; Arshad, R.; Cucchiarini, M. Nanomaterials for the Diagnosis and Treatment of Urinary Tract Infections. Nanomaterials 2021, 11, 546. [Google Scholar] [CrossRef] [PubMed]
  295. Abu Lila, A.S.; Huwaimel, B.; Alobaida, A.; Hussain, T.; Rafi, Z.; Mehmood, K.; Abdallah, M.H.; Al Hagbani, T.; Rizvi, S.M.D.; Moin, A.; et al. Delafloxacin-Capped Gold Nanoparticles (DFX-AuNPs): An Effective Antibacterial Nano-Formulation of Fluoroquinolone Antibiotic. Materials 2022, 15, 5709. [Google Scholar] [CrossRef] [PubMed]
Figure 1. E. coli with a representative set of virulence factors of the UPEC pathotype.
Figure 1. E. coli with a representative set of virulence factors of the UPEC pathotype.
Microorganisms 11 02169 g001
Figure 2. Cell targets of antibiotics used to treat infections by UPEC.
Figure 2. Cell targets of antibiotics used to treat infections by UPEC.
Microorganisms 11 02169 g002
Table 1. Treatment guidelines according to the European Association of Urology for Uncomplicated and Complicated UTI.
Table 1. Treatment guidelines according to the European Association of Urology for Uncomplicated and Complicated UTI.
Recommended First-Line Treatment Recommended Empirical Treatments
Uncomplicated cystitisFosfomycin trometamol (Oral)Trimethoprim
NitrofurantoinTrimethoprim/sulfamethoxazole
Cephalosporins (alternative)
Pivmecillinam
Uncomplicated pyelonephritisFluoroquinolonesCiprofloxacin
Levofloxacin
Trimethoprim/sulfamethoxazole
Cefpodoxime or Ceftibuten
Complicated UTIsAmoxicillin plus an aminoglycoside3rd generation cephalosporin used intravenously
2nd generation cephalosporin plus an aminoglycoside
The guidelines in Table 1 are taken from the European urology association “Guidelines on urology infections 2023” [7]. First-line therapy refers to the initial recommended antibiotics for treatment of patients and are chosen with considerations given to antibiotic susceptibility results. Empirical treatment refers to antibiotics recommended during the period prior to the receipt of antibiotic susceptibility results. Uncomplicated UTIs are defined as acute, sporadic, or recurrent lower (uncomplicated cystitis) and/or upper (uncomplicated pyelonephritis) UTIs, limited to non-pregnant women with no known relevant anatomical and functional abnormalities within the urinary tract or comorbidities. Complicated UTIs include infections involving men, pregnant women, patients with relevant anatomical or functional abnormalities of the urinary tract, indwelling urinary catheters, renal diseases, and/or with other concomitant immunocompromising diseases.
Table 2. Adhesions and toxins in UPEC with associated genes and host interactions.
Table 2. Adhesions and toxins in UPEC with associated genes and host interactions.
AdhesionsGenesHost Receptor
Type 1 FimbriaefimABCDEFGHI [72]D-mannose-containing receptors, uroplakins [23]
P FimbriaepapABCDEFG [84]mannose-resistant binding to globoseries glycosphingolipids [24]
Afa/DrafaABCDEF [96]decay-accelerating factor, carcinoembryonic antigens [100,101]
S FimbriaesfaABCDEFGHS [149]Sialo-glycoproteins [89,92]
Type one secretion AtosRCBDAEF [106]human kidney and epithelial cells [106]
ToxinsGenesEffect in host
α-hemolysinhlyCABD [125,126] PBK and ACLY inhibition, GM-CSF upregulation [34,127,128]
Cytotoxic necrotizing factorcnf1 [136]Inhibits Rho family of GTPases [129]
Serine protease autotransportersat [146]Induces vacuolation in glomeruli and proximal tubules, degrades complement proteins [36,147]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Whelan, S.; Lucey, B.; Finn, K. Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment. Microorganisms 2023, 11, 2169. https://doi.org/10.3390/microorganisms11092169

AMA Style

Whelan S, Lucey B, Finn K. Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment. Microorganisms. 2023; 11(9):2169. https://doi.org/10.3390/microorganisms11092169

Chicago/Turabian Style

Whelan, Shane, Brigid Lucey, and Karen Finn. 2023. "Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment" Microorganisms 11, no. 9: 2169. https://doi.org/10.3390/microorganisms11092169

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