Next Article in Journal
High-Energy Excimer Annealing of Nanodiamond Layers
Previous Article in Journal
Eco-Friendly and Sustainable Pathways to Photoluminescent Carbon Quantum Dots (CQDs)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanotechnology Involved in Treating Urinary Tract Infections: An Overview

by
Andreea Crintea
1,
Rahela Carpa
2,*,
Andrei-Otto Mitre
3,
Robert Istvan Petho
4,
Vlad-Florin Chelaru
4,
Sebastian-Mihail Nădășan
4,
Lidia Neamti
1 and
Alina Gabriela Dutu
1
1
Department of Medical Biochemistry, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
2
Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
3
Department of Pathophysiology, Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
4
Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Nanomaterials 2023, 13(3), 555; https://doi.org/10.3390/nano13030555
Submission received: 11 January 2023 / Revised: 22 January 2023 / Accepted: 27 January 2023 / Published: 30 January 2023
(This article belongs to the Section Biology and Medicines)

Abstract

:
Considered as the most frequent contaminations that do not require hospitalization, urinary tract infections (UTIs) are largely known to cause significant personal burdens on patients. Although UTIs overall are highly preventable health issues, the recourse to antibiotics as drug treatments for these infections is a worryingly spread approach that should be addressed and gradually overcome in a contemporary, modernized healthcare system. With a virtually alarming global rise of antibiotic resistance overall, nanotechnologies may prove to be the much-needed ‘lifebuoy’ that will eventually suppress this prejudicial phenomenon. This review aims to present the most promising, currently known nano-solutions, with glimpses on clinical and epidemiological aspects of the UTIs, prospective diagnostic instruments, and non-antibiotic treatments, all of these engulfed in a comprehensive overview.

1. Introduction

Except for the urethra, the urinary tract remains uncolonized, although occasional contamination of bladder urine is possible, especially in women due to the short and direct trajectory of the urethra [1,2]. In order of importance, the antimicrobial barriers of the urinary tract are: continuity of the uroepithelium doubled by mucopolysaccharides, a glycosaminoglycan that antagonizes the adhesion of bacteria lacking special ligands, washing of the mucosa through the normal urine flow (with a minimum dead space by complete evacuation of the bladder during normal urination), high osmolarity and high concentration of urea as well as a low pH value of urine, and the antibacterial effects of prostate secretion and periurethral glands [3,4,5,6,7].
Working synergistically, all these factors mentioned above create an unfavorable environment for the development of many bacteria in the urinary tract. However, like in the proximal parts of other orifices, the distal urethra is normally colonized with bacteria from the perineal skin, colon, and vulva. Thus, in the distal urethra, in moderate amounts, coagulase-negative staphylococci, Enterobacteriaceae, enterococci, Fusobacterium spp., Gram-positive and Gram-negative anaerobic cocci, lactobacilli, or even Candida species are very common [8]. Eventually, this resident microbiota also plays a discrete role in maintaining a proper barrier against threatening pathogens, establishing both fine and fascinating lines between what we can call ‘bad’ and ‘good’ germs [9].
Urinary tract infections (UTI) represent an enormous health problem globally, affecting both sexes, regardless of age, and they can vary in symptomatology and prognosis, being complicated or uncomplicated [10].
The diagnosis of UTI is done based on the reporting of symptoms that are supported by laboratory tests represented by urinalysis and urine culture [10]. The microbiological diagnosis of urinary infections is made by isolating pathogenic organisms, for a positive diagnosis of UTI [11]. The antibiogram helps to check the effectiveness of the therapy in fighting the infection. Urine culture is not mandatory in patients who do not present risk factors for developing a complicated UTI, but with its help the possible development of antibiotic resistance of uropathogens is reduced [10].
Catheter-associated urinary tract infections have significant clinical and economic consequences. Catheter-associated bacteriuria may be associated with excess mortality, even after controlling for underlying factors such as severity of illness and comorbidities; hospital-onset bloodstream infection resulting from a urinary source has a case fatality of 32.8% [12].
European studies found that 15–25% of patients in hospitals and 5% of those in elderly homes have been subjected to urinary catheter insertion. Further studies estimate that 41–58% of existing catheters are most likely needless. The risk to acquire CAUTI increases by 5% with daily catheter use. The CAUTI annual cost in the UK is over EUR 100 million annually (per episode it is over EUR 2000) (European Centre for Disease Control (ECDC) and European Network for Safer Healthcare (ENSH)).
Data from ECDC and ENSH help us to know that 6.5% of hospitalized European patients suffered a healthcare-associated infection and about 19% to 20% had a UTI. Urinary tract infections are one of the three most common medical infections. Catheter-associated urinary tract infections also mean an additional EUR 390 million in annual costs for the EU, and also around 400,000 additional hospital days, equaling around 3800 life years.
With around 400,000 hospitalizations annually just in the United States and a burdening cost of around USD 2.8 billion for the healthcare system [13], it is clear that UTIs could be managed through more refined approaches, considering their current large spread and financial footprint. The notably increasing incidence of UTIs by 52% from 1998 to 2011 [13], together with a doubled cost per patient care in the same period [14], outlines the above-mentioned statement. Another interesting study revealed that EUR 13.5 million was spent only in France in 2012 for the treatment of suspected UTIs that eventually had negative cultures of urine [15]. This analysis strongly suggests that the diagnostic tools should also be considered for improvement and as a cost-cutting instrument. Finally, the rising threat of antibiotic resistance among infection cases in general and UTIs in particular has also led to somber predictions in terms of cost risings. A study from Lebanon shown that patients infected with resistant urobacteria have 29% higher hospitalization costs, not only because the aggravated forms of their conditions, but also because of a longer length of stay in the hospital [16]. In the USA, the healthcare system may be under even more pressure due to this phenomenon, since it may add USD 2.2 billion annually to the costs with UTI patients, which is roughly the total amount of money spent on the remedy of all UTI cases [17].
To sum up, there are many aspects of this type of infection that may influence not only their clinical outcome, but, implicitly, the cost involved in them, with the most significant one being represented by the bacteria’s resistance to antibiotics. Considering the almost ubiquitous incidence of UTIs and the already-known economic burden generated by this condition for the national health services, it is our conviction that more experimental studies and reviews addressing novel approaches in the management of these conditions should be encouraged, published, and highlighted.

2. Clinical-Epidemiological Considerations

The urinary tract infection may be inapparent or overt. It affects any segment of the urinary tract and, due to the anatomical relationships, once installed in one segment, it extends to the others, leading to related diseases: urethritis, prostatitis, cystitis, and pyelonephritis [18]. In view of its manifestations, they generally range from episodic, limited acute episodes to recurrent, chronic conditions. Regarding its localization, kidney limitation can be a constant stage, as in leptospirosis, or occasional, such as in pyelonephritis with Candida albicans, Haemophilus influenzae, staphylococcal renal abscesses, renal tuberculosis, and so on. On the other side, renal elimination of the bacterium is achieved without clinically manifested kidney damage [19]. Finally, it is worth being mentioned that neither serum antibodies (IgM, IgG) nor those excreted in the urine (IgG, IgA) seem to play a role in kidney defense. In inferior urinary tract infections, the immune response is absent, and the infection is largely superficial [20].
Regarding the most affected age categories, urinary tract infections are relatively frequently diagnosed at all ages. However, due to a less efficient immune function, cumulative comorbidities, or increased risks related to nosocomial infections, the elderly have higher chances to develop symptomatic UTIs [21,22]. In newborns and infants, the prevalence is higher in boys than in girls. Subsequently, the ratio reverses more and more sharply until adulthood. However, the difference between sexes disappears in the elderly, due to the faster increase in the prevalence in men related to prostate diseases and the instruments they require. It is also worth mentioning that about 10–20% of women experience urinary tract infections at least once in their lifetime [23].
Even if we mentioned above several anti-infective barriers of the urinary tract, these anatomo-functional components still have various degrees of instability. For example, unlike other tissues, on the bladder mucosa, the phagocytosis of surfaces is practically absent [24,25]. Furthermore, residual bacteria can multiply in the urine film in the interval between urination, while complement effector and phagocytosis are useless in the hypertonic conditions of the renal medulla [25,26,27]. Different conditions may modify the balance of the microorganism and host to the detriment of the host, with the appearance of colonization or urinary tract infections. Obviously, the ascending path is the most important for conditioned bacterial pathogenic access in the urinary tract [28].
From a microbiological perspective, Escherichia coli is the most common etiological agent of urinary tract infections. Only a few serogroups of Escherichia coli have uropathogenic abilities, such as: recognizing sphingolipid receptors on the uroepithelium, the abundance of the K antigen, and resistance to the bactericidal action of blood serum or hemolysin production. Other serogroups, although predominant in the colon microbiota, produce only occasionally abortive urinary colonization or cystitis [29,30,31].
The rest of the Enterobacteriaceae, enterococci, or Pseudomonas aeruginosa cause chronic or recurrent infections; due to obstructive uropathy and neurogenic bladder dysfunction, Staphylococcus saprophyticus is a more common cause of cystitis in sexually active young women [32,33]. Corynebacterium urealyticum has recently been reported to cause urinary tract infections in patients at risk for hospitalization, prolonged antibiotic treatment, or urological interventions. Candida albicans causes specific infection in metabolically uncontrolled diabetics [34,35]. Urease-positive species, like P. mirabilis and S. saprophyticus, alkalize urine and promote the production of stones, which creates major difficulties in eradicating certain pyelonephritis [36].
Concerning the viral UTIs, serovar adenoviruses 11 and 21 cause hemorrhagic cystitis in children, but, in general, viruses that are excreted in the urine do not cause urinary tract infections [37].

3. Antibiotics

Chemotherapeutic substances are unknown products to the body which suppress or damage tumor cells and microorganisms [38,39,40]. Among them, we can mention antibiotics, antifungals, and so on. Antibiotics are substances with antibacterial effect, effective on bacteria in low concentrations, which observe the principle of selective toxicity, acting at the molecular level on an essential metabolic pathway for bacteria; the metabolic pathway being absent in the eukaryotic cell, or, if present in eukaryotes, not being affected by the antibiotic [41,42,43,44].
The modern era of antibiotics began with the discovery of penicillin by Sir Alexander Fleming in 1928 [45]. Even at the time, it was a revolutionary medicine; penicillin resistance became soon a critical issue and new antibiotics began to be discovered, developed, and implemented. Thus, from the late 1960s to the early 1980s, the pharmaceutical industry introduced new antibiotics to the market to solve the problem of resistance of various microorganisms to antibiotics [46,47,48,49].
In 2015, many decades after the first patients were treated, infections caused by microorganisms became a new threat [49,50]. Antibiotics fight bacteria by inhibiting certain vital processes of bacterial cells or bacterial metabolism [51,52] and can target the bacterial wall or cytoplasmic membrane or localized processes in the bacterial cytoplasm such as protein synthesis, DNA replication, or both [51,53,54]. For each of the groups listed above, the classification into families is based on the chemical structure of the various substances, with each family having a specific molecular mechanism [55,56].
Antibiotic resistance is an important and topical issue, which refers to the situation where antibiotics that usually kill bacteria no longer do so [55]. The study of antibiotic resistance has focused especially on the consequences of resistant microorganisms, as it can interfere with the treatment of infections [57,58]. Thus, it was demonstrated that bacteria were able to develop resistance, not only because the acquisition of antibiotic-resistant genes, but also as a result of excessive and abusive use of drugs and the lack of new drugs [49,59]. In consequence, patients infected with resistant bacteria will manifest symptoms for a longer time, and the chances of the conditions getting worse will be higher. Moreover, epidemics will be on the rise and more and more people will be at risk of infection [60,61,62,63].
The main causes of antibiotic resistance are their incorrect use: either empirical treatment is performed (without performing an antibiogram to test germ sensitivity); strong antibiotics are prescribed for infections that could be treated with simple antibiotics; or administration is in too small amounts, for too short a period, or at too long intervals [59,64]. The most worrying cases are for the species of the family Enterobacteriaceae—Escherichia coli and the Klebsiella genus.
Resistance of microorganisms to antibiotics can be natural or acquired. The natural resistance of a species or genus is a characteristic of its own [65,66,67]. It is always transmissible to offspring because it is carried by the chromosome. Thus, natural resistance determines the wild phenotypes of bacterial species against antibiotics [68,69]. Unlike natural resistance, acquired resistance refers only to a proportion of strains belonging to a species or genus, variable over time. Acquired resistance exists through the accumulation of one or more resistance mechanisms that determine a certain resistance phenotype, a different phenotype from the wild one [70,71,72]. Among the mechanisms for obtaining antibiotic resistance, we mention the genetic mechanisms for acquiring antibiotic resistance and the biochemical mechanisms. As seen in Figure 1, the change in the permeability of the bacteria to antibiotics occurs through mutation [67,73].
Antibiotics enter the Gram-negative bacteria through the pores or lipopolysaccharide of the outer membrane [74]. Changing the permeability of bacteria by mutation, by changing porins or lipopolysaccharides, prevents the penetration of antibiotics into the bacteria [75,76,77]. By changing the permeability of bacteria to hydrophilic or hydrophobic antibiotics, co-resistance or cross-resistance is installed against a family of antibiotics, a group of antibiotics with the same physicochemical properties [74,78]. By changing the permeability, resistance to various families of antibiotics can be acquired: beta-lactams, tetracyclines, fennel, polymyxins, and so on [79,80,81]. Modification of antibiotic transport in bacteria occurs by mutation, meaning that the antibiotic can no longer be transported in bacteria. Other biochemical mechanisms for acquiring antibiotic resistance that may be mentioned are the modification of the antibiotic action target, the substitution of the action target, the expulsion of the antibiotic, or the inactivation/modification of the antibiotic by inactivation enzymes (Figure 2) [82,83,84]. From the last category mentioned above, it can be said that the inactivation or modification enzymes of some antibiotics generally have a plasmid and rarely chromosomal determinism and can act either in the extracellular or intracellular periplasmic space of bacteria (beta-lactamases in Gram-negative bacteria) [70].
Finally, a prediction generated in 2016 estimated that more than 10 million people will die annually by 2050 because of the antimicrobial resistance (AMR) phenomenon [85]. Corroborating the fact that UTIs account for a great percent of all these infections and the fact that, recently, there has been a clearly rising AMR trend, especially among these kinds of infections [86,87] outlines the need for novel and more efficient clinical approaches regarding the management of UTIs. With an inefficiency already varying from 32 to 60% in terms of antibiotic medications around the globe [86], it seems that classical therapy is more and more suboptimal for the clearance of infections.
With these brief statistics in mind, we chose to present a complete picture on how the antimicrobial medication evolved, rather than just enumerating novel technologies used for urinary infection healing. Starting from basic, primary therapeutics and expanding to the current state of research on UTIs treatments, in the following chapter we extensively detail the nanotechnology’s involvement and their perspectives in this clinical topic.

4. Nanotechnology Used as a Diagnostic Method and in Antimicrobial Treatment

Diseases caused by various pathogens are numerous and can be easily transmitted. Rapid and specific determination of pathogens is particularly important to determine the source of infection and the spread of infection but also the proper treatment of patients [88]. The complexity and variety of pathogens, and also the incubation period until the onset of the first symptoms of the pathology, can make the diagnosis difficult. Modern diagnostic methods such as ELISA and PCR are characterized by high sensitivity and reproducibility. These methods also have disadvantages: laborious preparation of samples and long time until conclusive results are obtained [89].
Nanotechnology presents a great opportunity for the development of fast techniques, characterized by high sensitivity, specificity, and low costs for the detection of infections caused by various pathogens (Figure 3) [90,91,92].
Various types of nanomaterials, such as magnetic and fluorescent gold nanoparticles, are used to determine microorganisms. The versatility of these inorganic nanoparticles, together with the interactions with different target molecules or pathogens, makes their use promising for diagnosis [92,93,94,95]. It should be noted that these diagnostic strategies, which are based on nanoparticles, depend on the recognition of different sequences in the bacterial genome and that is why no new strains can be identified [96]. Resistant strains continue to emerge, and nanotechnology needs to provide new ways to identify both the presence of pathogens and the sensitivity of pathogens to antibiotics [97]. There is great interest in the rapid development of a diagnostic method for urinary tract infections. As early as 1980, various methods began to be developed to help detect urinary tract infections—for example, immersing a strip in urine to assess the esterase produced by leukocytes and nitrites [98]. More recently, automated analyzers that are based on the principle of flow cytometry allow rapid detection of bacteria, leukocytes, erythrocytes, epithelial cells, crystals, and so on. Although this method is quite fast, it cannot provide the microbiological diagnosis nor the susceptibility of microorganisms to different classes of antibiotics [99]. The standard procedure for detecting urinary tract infections is based on several steps that include the collection and transport of the biological sample. Once the results are available, each clinical laboratory has to notify the clinician about the result of the analysis, who in turn informs the patient about the result obtained [100,101]. All these steps that need to be taken are time-consuming and can delay the provision of effective treatment. The new generation of biosensors based on micro- and nanotechnologies offers the possibility of a very efficient molecular diagnosis [102,103]. A biosensor is a device consisting of the combination of two main elements: a sensitive, biological element, the bioreceptor, which, immobilized on a support, can specifically recognize a target substance present in a complex environment; and a physicochemical system, which captures the changes in the bioreceptor in contact with the target substance and translates them into a useful electrical signal [104,105]. The bioreceptor can be of the catalytic (metabolic) type, such as an enzyme, a cell organelle, a microorganism, or even a tissue, but also of the non-catalytic (affinity) type, such as an antigen or an antibody [106,107]. In developing a biosensor for urinary tract infections, several aspects need to be considered. To perform the analyses of interest in the case of urinary tract infections, several steps are required: sample preparation—pipetting, centrifugation, and washing. The sensitivity of the biosensor can be determined by several factors, such as matrix effects and nonspecific binding [100,108]. A urinary tract infection sensor should meet several criteria: the ability to detect infection, quick performance of the analysis, the automatic preparation of the sample with minimal technician intervention, the ability to identify pathogens, and antimicrobial sensitivity, so as to be versatile enough to be able to identify different pathogens in different infections [100]. The emergence of antibiotic resistance of microorganisms has become a threat to human health. The study of antibiotic resistance has focused on pathogenic microorganisms, but also on the consequences that resistant microorganisms have on health [58,109]. The development of antibiotic resistance is very important from a clinical point of view, as it can be detrimental to the treatment of infections. Shortly after the introduction of antibiotics for human therapy, bacteria were able to develop resistance, as a consequence of mutations, the acquisition of antibiotic-resistant genes, and the overuse and abuse of drugs, in addition to the lack of new drugs [59]. The natural resistance of a species or genus is a characteristic of its own. It is always transmissible to offspring because it is carried by the chromosome. Thus, natural resistance determines the wild phenotypes of bacterial species against antibiotics. Unlike natural resistance, acquired resistance refers only to a proportion of strains belonging to a species or genus, variable over time [110]. Acquired resistance exists through the accumulation of one or more resistance mechanisms that determine a certain resistance phenotype, a different phenotype from the wild one [111]. There are three categories of mechanisms responsible for the acquired resistance of bacteria to antibiotics. The first one is represented by a reduction in the amount of antibiotic that reaches the target by decreasing permeability or by the appearance of efflux systems [112]. Decreased permeability is found especially in Gram-negative bacteria, in which the pores are partially or completely blocked or may disappear. The crossing can also be slowed down due to mutations, so that the accessibility of antibiotics to the pores outside the outer membrane decreases, and using outflow systems that use a proton-motor force that expels the antibiotic from the moment it enters the bacterial cell [113,114].
Other different mechanisms responsible for the acquired resistance of bacteria to antibiotics are represented by the modification of the antibiotic target, which can be achieved by mutations in the genes encoding the antibiotic target, by acquiring foreign genes, or by the inactivation of the antibiotic, which is the most common mechanism encountered in infectious pathology [73,112]. Of the resistance mechanisms, one of the important ones is the expression of enzymes that degrade or modify antibiotics, such as β-lactamases, which remains one of the significant threats against antibiotic efficiency [42].
Nanomedicine plays an important role in enhancing the effectiveness of existing therapies through increasing the stability and the physicochemical properties of antibiotics and antibiotic release and offering an opportunity for biofilm internalization, further increasing the capability for target delivery of drugs to the infection site [115,116,117]. It was also found that, in addition to improving the therapeutic activity of antibacterial agents, nano-sized systems also restrain the stimulation of resistance through overcoming the developed resistance strategies of the bacteria [118,119]. These strategies involve drug decomposition by β-lactamase or efflux pumps or modification of the bacterial cell wall. As most types of nanoparticles can overcome at least one of these common resistance mechanisms, an increased number of nanoparticle variants have been used as an improved line of defense against antibacterial resistance [120].

4.1. Organic Nanoparticle Therapy Approaches

The overall advantages lately brought by the usage of organic nanoparticles are already well known. Starting from a more precise targeting of various tissues/environments or even cells and going to a strictly controlled on-site dosage and enhanced colloidal stability of small drugs (like proteins or nucleic acids), these nano-sized carriers revolutionized the concept of modern therapy (Figure 4) [121,122].
In the case of catheters and other traditional medical devices/approaches used in treating urinary diseases, a great risk for the patient is that of developing biofilms (Figure 5) [123,124].
In the light of this issue, we present several promising findings related to the usage of nanoparticles in UTIs therapy management.
The first example refers to a food sweetener naturally present in ginger called zingerone [125]. Zingerone nanoparticles (ZNPs) were tested in vivo on mice as an alternative to standard therapies against Pseudomonas aeruginosa, one of the bacteria able to produce biofilms [126]. After treatment, both mice were treated with zingerone and with ZNPs, showing a decreased bacterial count in both renal and bladder tissues compared to untreated controls. Moreover, while controls showed increases in some inflammatory markers (MDA, MPO, and reactive nitrogen intermediates), mice treated with zingerone or ZNPs showed decreases in those markers, ZNPs having a stronger effect than zingerone itself. Lastly, ZNPs increased serum sensitivity and phagocytosis of Pseudomonas aeruginosa compared to zingerone, which summarizes perfectly the importance of considering nanoparticle formulations rather than classical, simple drugs for the treatment of UTIs [127,128].
Nanocarriers were also considered as remedies for UTIs in the light of improved bioavailability and better retention in the target tissue. For example, Brauner et al. [129] proposed a formulation of Trimethoprim (TMP) as poly(D,L-lactic-coglycolic acid) (PLGA)-based nanospheres, further conditioned with wheat germ agglutinin (WGA), to be administered intravesically through a catheter [129,130,131]. TMP is used to treat uropathogenic Escherichia coli infections, and while usually TMP is administered orally, there have been other cases of medicine administered intravesically because of the increased local concentration and to avoid absorption losses or systemic effects [132]. Moreover, WGA is used to mimic the role of Escherichia coli type 1 fimbriae (FimH), both having increased binding activity for the bladder epithelial tissue [133]. PLGA-coated TMP nanospheres, with and without WGA added to the surface, were tested on immortalized human uroepithelial cells. On repeated washings of the urothelium after the initial application of the formulations, the authors noted higher adhesions for nanoparticles of higher WGA density. Overall, there was no visible connection between TMP loading and nanoparticles adhesion. Lower pH levels and higher incubation times favored the adhesion of the nanoparticles to the urothelium [129,134].
As antibiotic resistance was already identified as a major worldwide concern, solutions involving organic nanoparticles can also be used to prevent antibiotic resistance in target bacteria. For example, Amikacin (AK) resistance can be gained through multiple mechanisms (mutations of the 30S subunit of the ribosome, aminoglycoside-modifying enzymes (AME), changes in lipid content of the plasma membrane, efflux pumps) [73,135,136]. Moreover, antibiotics like Amikacin have narrow therapeutic indexes and strong systemic countereffects. In such cases, nanocarriers can be used to improve bioavailability and target specific tissues [137]. As a solution for this issue, scientists designed liposomes that can entrap a range of antibiotics (including Amikacin) which are then able to fuse with the bacteria’s membrane and deliver their content directly into the antibiotic-resistant bacteria. In this way, on the outside of the cell, the antibiotic molecule is protected from AME action by the liposome vehicle [138,139].
Additionally, if the above-mentioned strategy is still insufficient for the desired results, it is also worth mentioning that surfactants such as Poloxamine 908 are able to influence the uptake in desired tissues (decreasing the liver uptake while increasing the spleen uptake) [140]. Some polymeric nanoparticles are also able to increase uptake in a specific tissue, further helping to target the desired infection location (such as Amikacin-bearing PLGA nanoparticles that have an increased uptake in Peyer’s patches compared to plain Amikacin) [141].
Going even further, considering that the localization of UTIs may sometimes be hard to determine, Song et al. [142] created a copolymer (LGseseTAPEG) of PLGA, selenocystamine, and methoxypoly(ethylene glycol) tetraacid which is sensitive to local oxidative stress [142].
Besides being effective drug carriers, organic nanoparticles can also be used as detectors of pathogens. One example of a nano-sized UTI detection system is Cranberry proanthocyanidins (PAC), which can interact with fimbriae of extra-intestinal Escherichia coli [143]. PAC-polyaniline (PANI) nanocomposites were created and used on screen-printed electrodes (SPE) to assess electrochemical changes caused by the interaction between PAC and Escherichia coli [144,145]. The resulting PAC-PANI nanocomposites on SPE produced a linear electrical response towards Escherichia coli presence over the range of 1 to 70,000 colony-forming unit/mL (CFU/mL), and the limit of quantification was 1 CFU/mL, much lower than earlier approaches in the literature. Moreover, the electrical response was different for other pathogens, and as such the approach was also specific for Escherichia coli [144].
Finally, in a similar manner, nanoparticles may be used to create noninvasive, urine-based tests for diseases that are not specific to the urinary system [146]. In a clinical study, HIV-positive patients presenting neurological symptoms specific for toxoplasmic encephalitis (TE) were tested both through qPCR and through NPs of poly-N-isopropylacrylamide dyed with reactive blue-221 [147,148]. The NPs, which captured and concentrated relevant antigens from urine, were then evaluated through western blot immunoassay. Unfortunately, due to the lack of a golden standard test (qPCR has a sensitivity of under 70%), the authors were unable to compute the sensitivity and specificity of the NP-based technique described [147].

4.2. Inorganic Nanoparticle Therapy Approaches

As a more stable and less toxic alternative, inorganic nanoparticles may be used to respond even better to the challenges that emerge in the therapy of UTIs. Additionally, considering that many of these NPs have antibiotic properties themselves, reducing bacteriuria in infections caused by both multi-drug resistant (MDR) and common pathogens, they may be especially recommended for the treatment of UTIs [131]. Thus, we try to present the usage of inorganic NPs in UTI management in a more comprehensive manner.
In the search for effective and new ways to treat urinary tract infections caused by MDR pathogens, biogenic selenium nanoparticles (Se NPs) have been created through two different methods: by biogenic synthesis using Penicillium chrysogenum filtrate and by incorporating Se with Gentamicin (CN) under gamma radiation (green synthesis) [149]. The Se NPs incorporated with CN (Se NPs-CN) exhibited antimicrobial activity against Escherichia coli, Staphylococcus aureus, Bacillus subtilis, Pseudomonas aeruginosa, and also towards Candida albicans. The biogenic Se NPs showed effectiveness against Staphylococcus aureus, Candida albicans, and Pseudomonas aeruginosa [150,151,152]. The antimicrobial activity was more significant compared to the precursor sodium selenite, the fungal filtrate, Gentamicin alone, and other standard antimicrobial agents. Additionally, the Se NPs-CN presented a more efficient activity compared to the biogenic Se NPs [151,153]. The Gram-negative bacteria were more susceptible to the Se NPs action compared to the Gram-positive. It was also observed that Se NPs-CN had antibiofilm activity against Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus [154,155]. The mechanism of the Se NPs is based on the fact that selenium is capable of producing highly effective superoxide radicals which create a thiol deficiency in the bacteria, which causes cell death (Figure 6). It is also important to note that a dose higher than 1.8 mg/kg produced chronic hazardous effects on tested mice [151,156].
Since metal NPs do not aim to induce alterations in metabolic pathways, they cannot trigger the bacterial resistance mechanisms, so these agents can be considered excellent candidates for the treatment of UTIs. Still, the production process of these instruments may have detrimental effects on the environment and raise questions with regard to the side effects manifested in the patients [36,131,157,158]. To overcome these concerns, M. Abd Elkodous et al. [159] studied zinc oxide nanoparticles (ZnO NPs) produced in an eco-friendly and cost-effective manner through the sol-gel method [159]. The nanoparticles were tested against multidrug-resistant bacteria involved in UTIs. The ZnO NPs presented high thermal and chemical stability, but also anti-corrosive properties. The manufacturing process (the sol-gel method) is quite simple and it provides pure particles [160,161]. The synthesized ZnO NPs showed antimicrobial activity against multiple UTI pathogens. They was most effective against Bacillus subtilis, but also effective against Pseudomonas aeruginosa and fungi such as Candida tropicalis. The ZnO NPs presented much higher antimicrobial activity than zinc nitrate or other antimicrobial agents [162,163,164]. It was also noticed that the Gram-negative bacteria are more sensitive to the ZnO NPs than the Gram-positive ones. The ZnO NPs also possess very good surface area, low crystal size and good porosity and have an anisotropic nature; all these characteristics allow the ZnO NPs to be more effective. The antibiofilm activity was presented against Bacillus subtilis, Pseudomonas aeruginosa, and Candida tropicalis. The ZnO NPs inhibit the formation of biofilm in the initial stage, also named the irreversible adhesion stage. The ZnO NPs hinder the production of exopolysaccharides, which in the end leads to the inability of Bacillus subtilis to form the biofilm [165,166]. It was also observed that the ZnO NPs had some antitumor properties; more precisely, there was an in vitro cytotoxic effect against Ehrlich ascites carcinoma (EAC). The ZnO NPs’ antioxidant potential was also confirmed through the continual reduction of zinc ions. The ZnO NPs were proven to be effective in the treatment of UTIs and fungi infections and, even further, showed great antitumor action [167,168]. Due to these particular advantages, it would be worthwhile to pursue further research of its properties and applications in industries such as food processing, pharmaceuticals, and cosmetics, in which the ZnO NPs could have a positive impact [160,169,170].
In an attempt to find new ways to diagnose UTIs in the early stages, S. Vasudevan et al. developed a photoluminescence biosensor based on ZnO Cysteamine functionalized nanoparticles [171]. This type of sensor has a fast response time, high sensitivity, and specificity. The ZnO NPs combined with Cysteamine (ZnO-Cys) have shown maximum sensitivity towards detection of N-Acyl-Homoserine Lactones (AHL) produced by Gram-negative bacteria, such as Pseudomonas aeruginosa. The AHL is used by the Gram-negative bacteria as a signaling molecule for intra-species and also inter-species communication [172,173]. For example, Escherichia coli can sense and detect AHL produced by other bacteria, which leads to the regulation of virulence of the pathogen. The ZnO NPs have photoluminescence properties and, with the help of Cysteamine, the nanoparticles can be used in binding and detecting AHL molecules [174,175]. The specificity of the nanoparticles was demonstrated by using an artificial urine medium (AUM) which contained the necessary components to be as similar as possible to human urine. Pseudomonas aeruginosa was also added to the AUM, and, in this way, it was possible to have a real-time detection. The ZnO-Cys detected AHL molecules after 30 min and reached a maximum of detection at 90 min, after which point there was a decrease in intensity [173].
Another study performed by Muhammad Ali Syed et al. [176] manifested a particular interest in tungsten nanoparticles [131,177]. As UTIs are one of the most frequent nosocomial infections, with most of them being caused by improper catheter usage, the cited experiment comes in response to this well-known origin of these infections. As multiple drug-resistant (MDR) pathogens, especially Escherichia coli, are responsible for many of the catheter-associated urinary tract infections (CAUTIs), we will outline the importance of such studies in the context of CAUTI therapy management [178,179]. The Tungsten nanoparticles (W-NPs) were tested against the MDR Escherichia coli isolated from hospitalized patients and its efficiency was compared to Cefotaxime sodium and Sulfamethoxazole. The Escherichia coli isolated in this study were resistant to both antibiotics and also presented different genes which encoded adhesins and toxins. The W-NPs, when compared to the antibiotics, showed a higher efficiency at a lower MIC (minimum inhibitory concentration) and it was clear that the W-NPs determined growth inhibition, most probably because of the accumulation of intracellular reactive oxygen species [131,176]. One of the solutions for CAUTI would be the incorporation of antimicrobial agents such as W-NPs in the catheter itself. Due to the W-NPs’ nonspecific way of action, the risk of developing bacterial resistance is much lower and its design could stop the production of biofilm. Another advantage of the W-NPs in comparison to silver, for example, is the lower toxicity [180,181]. However, these nanoparticles were rather poorly studied in the context of CAUTI treatment alternatives, since there are no other relevant mentions in the literature on this issue. This fact highlights the significant perspectives posed by the development, testing, and implementation of therapies based on W-NPs [131,182].
Ureteral stent-related urinary tract infections are another issue that may be solved through the use of inorganic nanoparticles. The rate of colonization on the ureteral stents can be between 42–90%, depending on the study [183]. Not only is there a greater risk of developing a UTI, but the extraction of the stent is painful and invasive. Having that in mind, Gao et al. [184] tried to create a ureteral stent with antibacterial properties which is also biodegradable so that it does not have to be removed [184]. The stent requires flexibility and other mechanical properties to sustain the pressure of the peristaltic contractions. The stent developed by Gao et al. [184] is made out of a hyperbranched poly (amide-amine)-capped Ag shell and Au core [184]. The Ag and Au NPs were chosen because of their effective antibacterial activity and also because of the very small amount of Ag released and negative cytotoxicity. With the help of the stent’s self-cleaning capabilities, the contact surface implanted with Ag@Au NPs is constantly renewed and the debris is also removed [185,186]. In vitro tests showed that the Ag@Au NP-embedded ureteral stents were effective against Escherichia coli (growth inhibition over 90%) and Sthapylococcus aureus (growth inhibition of 99.99%). With regard to the most important mechanisms of Ag@Au NPs, it is worth mentioning that the formation of reactive oxygen species and the interaction of Ag with enzymes and proteins in the bacteria are the most important ones, both leading to cell death. In vivo experiments confirmed its degradation properties, but also the fact that it was effective in preventing UTIs, supporting the promising perspectives of this medical device [29,184,187,188].
Furthermore, as the number of UTIs caused by MDR Escherichia coli is on the rise because of the massive use of broad-spectrum cephalosporins, another topic related to the use of nanoparticles in urinary tract infections was studied by Sajjad et al. [189]. Nowadays, there are strains of Escherichia coli which produce ESBL enzymes, a fact that leads to a significant drop in antibiotics’ effectiveness. These strains presented 100% resistance to Ceftriaxone, 94% against Ciprofloxacin, and 79% against Co-trimoxazole, so the team tried to find a solution for the MDR Escherichia coli [190,191,192]. Silver oxide (AgO2) nanoparticles have shown potential in treating such infections, so they were considered as the main candidates for this purpose. During the production of AgO2-NPs, silver foil was used as a cheaper precursor. The study showed that there is a positive synergy between Ceftriaxone (CRO) and AgO2-NPs, as this corroboration led to a significant increase in the size of the inhibition zone against MDR Escherichia coli. The antibacterial mechanism is based on an increase in membrane permeability and alteration of membrane transport. Thus, not only would this treatment be more effective, but it would also be less toxic, due to the lower dose of antibiotics [189].
Besides the wide range of therapeutic-oriented usages presented above, inorganic NPs can also be considered as valuable diagnostic tools. In this regard, Vaezi et al. [193] showed in a recent study the possibilities of using an effective and inexpensive NP-based diagnosis method for urinary tract infections [193]. Other studies have shown the application of different metal quantum clusters, which have fluorescent properties. Human hemoglobin contains iron quantum clusters (Hb-FeQCs) which can be used for bioimaging. The Hb-FeQCs can bind very easily to Cu2+ ions and that leads to turn-off of the fluorescence of the nanostructure. Escherichia coli possesses enzymes that help the bacteria bind and reduce the Cu2+. That is why, when Escherichia coli is present, it will remove the Cu ions from Hb-FeQCs, and in this way the metal quantum will regain its fluorescent property in 30 min. Therefore, it would be an easy method of identifying Escherichia coli strains in a rapid and effective manner even at a low Escherichia coli concentration of only 8.3 × 103 CFU/mL. The Hb-FeQCs are also water-soluble, stable, biocompatible, and highly sensitive to bacterial concentrations, especially in the urine medium [193,194,195].

4.3. Mixed Nanoparticle Therapy Approaches

Bringing together the advantages of both organic and inorganic NPs while also trying to avoid the drawbacks of each category, scientists tried to develop veritable high-yielding solutions to combat UTIs [196].
For instance, Gupta et al. [197] created Au-NPs functionalized with fluoroquinolone antibiotics, resulting in up to 16-fold decreases of the minimum inhibitory concentration of the antibiotic, both on Gram-positive and Gram-negative bacteria [197]. Much in the same way, Saha et al. [198] created Au-NPs conjugated with Ampicillin (AMP), Streptomycin, and Kanamycin (KAN) and tested their efficiency and stability on Escherichia coli, Micrococcus luteus, and Staphylococcus aureus [198]. Kanamycin Au-NPs had a stronger inhibitory effect on Escherichia coli and Staphylococcus aureus and Streptomycin had a stronger inhibitory effect on Micrococcus luteus. On the other hand, Staphylococcus aureus was resistant to Streptomycin, and as such neither free Streptomycin nor its Au-NP form managed to inhibit bacterial growth (Table 1). Overall, functionalized NPs also had stronger activity after being exposed to heat shocks or after being stored at room temperature, compared to their classic counterparts [199,200].
Another example of NPs made from combined materials is the hybrid silver-talc nanocomposites (Ag/Tlc NCs) created by Daghian et al. [201]. These complexes were also chitosan-capped (Ag/Tlc/Csn NCs) and performed impressively not only in the cytotoxicity and antibacterial activity tests but also on wound healing assessments in mice [201]. Standard talc preparation showed weaker antibacterial activity, while Ag/Tlc NCs and Ag/Tlc/Csn NCs were both stronger than the composites containing talc, but with no significant difference between themselves. Likewise, on the antioxidant activity tests, talc had a weaker effect, while Ag/Tlc and Ag/Tlc/Csn were on par with vitamin C for the same concentration. In a total tissue bacterial count, Ag/Tlc/Csn NCs showed the lowest count (highest antibacterial activity), which was significantly different from the Ag/Tlc or Tlc formulations [202].
Last but not least, we could not ignore the emerging niche of the mobile biosensors, which, based on an organic–inorganic conglomerate, interestingly found applicability in the treatment of UTIs as well [108,203]. Punctually, using a cellulose base assembled as origami in order to aid in compiling all the necessary analytical stages, antibody-decorated gold nanoparticles (Ab-AuNPs) are used to assess the presence of Escherichia coli in urine samples collected from anonymized patients. When a droplet of urine containing Escherichia coli is dried onto one fold of the paper and then transferred onto another fold containing Ab-AuNPs, the NPs change their color to red in a dose-dependent manner. Subsequently, a mobile app was developed and calibration was performed for a mobile phone to be able to quantitatively evaluate the Escherichia coli concentration by capturing the image of the Ab-AuNP loaded paper. Thus, it was proven that these biosensors may be helpful in the fast and cheap point-of-care diagnosis of UTI [204].
Table 1. Specific NPs approaches for treating UTIs.
Table 1. Specific NPs approaches for treating UTIs.
Organics NPsMicroorganismsActivityReference
KAN-chitosan NPsEscherichia coli, Proteus mirabilisAntibacterial[130,131,205]
NanodiamondsMicroorganismsActivityReference
NanodiamondsUPECAntibacterial[130,131,205]
Silver-based NPsMicroorganismsActivityReference
Silver NPsEscherichia coli, Staphylococcus aureusAntibacterial[130,131,205]
Silver NPs-AMPEnterococcus faecium, Staphylococcus aureus, Acinetobacter baumanii, Morganella morganii, Pseudomonas aeruginosa, Klebsiella pneumoniaeAntibacterial[130,131,205]
Silver NPs-AKEnterococcus faecium, Staphylococcus aureus, Acinetobacter baumanii, Morganella morganii, Pseudomonas aeruginosa, Klebsiella pneumoniaeAntibacterial[130,131,205]
Copper-based NPsMicroorganismsActivityReference
Copper oxide NPsMRSA, Escherichia coliAntibiofilm[130,131,205]
UPECAntibacterial[130,131,205]
Zinc-based NPsMicroorganismsActivityReference
Zinc oxide NPsCarbepenem resistant, Acinetobacter baumaniiAntibacterial[130,131,205]
UPECAntibiofilm[130,131,205]
Candida albicansAntifungal[130,131,205]
Candida albicansAntibiofilm[130,131,205]
Gold-based NPsMicroorganismsActivityReference
Gold NPsEscherichia coliAntibacterial[130,131,205]
Gold NPs-CHXKlebsiella pneumoniaeAntibiofilm[130,131,205]

5. Biocompatibility

Biocompatibility refers to the ability of NPs to perform their function without causing undesirable effects. The in vivo behavior is directly connected with the NPs’ physical properties [94,206].
In order to use the inorganic NPs in the biomedical field, there are two basic requirements which have to be met: water solubility and colloidal stability. In most cases, the use of hydrophobic NPs showed a poor dispersion in biological fluids and a tendency to form aggregates. Another essential condition would be to maintain the physical properties of the NPs even after entering the organism [207,208]. The NPs’ size influences the absorption but also the clearance of the particles. NPs measuring over 100 nm are isolated by the reticuloendothelial system; meanwhile, particles which are smaller than 5.5 nm, can pass the renal glomerulus easily [209]. Another factor on which the biocompatibility of inorganic NPs depends is the surface structure. The surface structure is determined by the type and density of decoration molecules. Different molecules will confer different properties and in vivo behavior [210]. The surface charge can also modify the way a NP behaves in vivo. The charge is totally conditioned by the coating molecules, which in the end interact with the serum proteins and the cell membranes, thus influencing the biocompatibility. It has been shown that a neutral surface presented minimum interaction with the serum proteins and a prolonged blood half-life [211,212]. The biocompatibility of inorganic NPs is also strongly correlated to the nano-bio interface and all the processes which take place at its level. The result of all interactions is called the ‘biomolecule corona’, with the protein part being the most researched (protein corona) [213]. This protein corona is able to modify the adhesion to cell membranes and the physicochemical properties and the pharmacokinetics of the inorganic NPs. Stealth NPs are currently being developed for optimal action in vivo, but more research is necessary [213,214].
Since it is still largely considered an emerging field, most nanoparticle studies have been done in vitro, and, as such, tissue targeting has been only poorly studied. For greater adoption among healthcare providers and patients, nanoparticles should have the necessary characteristics to be ideally delivered by oral intake [215].
Concerning the envelope composition, chitosan is regarded as the perfect drug carrier, both due to its ability to be processed when conjugated with various medicines (including antibiotics) and for being able to increase the oral bioavailability of other medicine. For example, chitosan is fully protonated at a pH of 4, thus enabling absorption in the gastric mucosa. Absorption in slightly basic media, such as the small intestine, is managed through thiolate chitosan and other variants. Moreover, curcumin-carboxymethyl chitosan can inhibit apical P-glycoprotein drug transporter, thus improving the drug absorption. Thus, chitosan-based NPs can be used for oral intake of drugs aiming for systemic effects or specific target tissues [216,217,218].
On the other hand, as already stated, NPs have the benefit of targeting specific tissue more than systemic-administered antibiotics. Thus, nanoparticles functionalized with antibiotics might result in lesser systemic effects than classic antibiotics [157]. Pertaining to cytotoxicity, an in vitro experiment on HEK-293 human embryonic kidney cells showed that, even at a concentration of 50 μg/mL, zingerone nanoparticles were less cytotoxic than zingerone itself (93.5% vs. 89% survival rate) [128]. Mixed silver, talc, and chitosan-based NPs were safe from a cytotoxicity standpoint, in an in vivo study, for concentrations of up to 0.50 mg/mL. Interestingly, the Ag/Tlc/Csn NPs have higher toxicity than talc itself, confirming the idea that silver is responsible for cytotoxicity [219].
Lastly, NPs were proposed for use in coating medical devices, such as Foley catheters. Oftentimes, Foley catheters are used for longer periods without being replaced. In this case, they become targets of bacteria colonization and biofilm creation, in which case NPs are used to inhibit bacterial activity [220]. Chlorhexidine-loaded nanoparticles (CHX-NPs) were layered on catheters, and then those catheters were prepared as thin films of 2.5 × 2.5 cm. Hemocompatibility was assessed through hemolysis and showed no statistically significant difference between coated catheters (with and without CHX-NPs) and the uncoated catheters. Moreover, both skin irritation and sensitization tests were negative; no clinical signs of sensitivity were observed, nor was there increased presence of granulocytes or lymphocytes in skin sections [221].

6. Conclusions and Future Perspectives

The current review bases its structure and content on the premise that UTIs represent one of the principal reasons for antibiotic usage nowadays. As this fact was frequently encountered throughout the documentation standing at the basis of this paper, we decided to create a path through all the innovations that came as alternatives to the classic therapies, involving plain gavage of antibiotics.
Even if the nanoparticle-based therapy approaches are regarded as very recent concepts, the current review highlights the great amount of research already made in this direction, a fact that confirms the high interest manifested for the substitutes of traditional medications given for UTIs. However, we can firmly state that nano-sized therapies developed so far to treat urinary infections do respect the general tendency to consider hybrid NP-antibiotics formulations rather than simply removing classical medications. The unquestionable advantage of this strategy is that, despite almost completely disregarding decades of antibiotics research, one can potentiate the effect of the currently available drugs by using them in new formulations.
With regard to the perspectives, the example presenting the Ag/Tlc/Csn NCs is a pertinent case of how scientists should tackle the problem of NPs’ cytotoxicity [201].
Besides their multiple advantages, we mentioned that inorganic particles can have major drawbacks related to toxicity. Thus, a strategy in which various types of metals or formulations are tested in the same therapeutic complex is highly advisable.
Furthermore, the unexpected presence of mobile biosensors in this niche clinical area cannot be overlooked. Starting as a question a decade ago, the development of such devices has become factual, the already-mentioned origami-based biosensor being able to detect the presence of Escherichia coli in urine samples in just 7 min. As the need for proper and in-time diagnosis of UTIs becomes more and more demanding, the interest for such user-friendly and effective mobile immunosensors will certainly rise [204].
Considering all the things mentioned before, there is certainly a great amount of effort maintained and fulfilled in the conquest of finding alternative cures for UTIs in order to avoid the antibiotic resistance triggers of pathogens. Nevertheless, the reluctance of patients to be given new trial formulations, the questionable safety of these complexes, and the hazards that may be caused by their production or disposal after usage are aspects that will be solved only with gradual, careful clinical implementation and supplementary research.

Author Contributions

Conceptualization, A.C. and A.G.D.; methodology, A.C., A.G.D., L.N., A.-O.M., V.-F.C. and S.-M.N.; validation, A.C., A.G.D., L.N. and R.C.; investigation, A.C., A.-O.M., V.-F.C. and S.-M.N.; resources, A.-O.M., R.I.P., V.-F.C. and S.-M.N.; writing—original draft preparation, A.C., A.G.D., A.-O.M., R.I.P., V.-F.C., L.N. and S.-M.N.; writing—review and editing, R.C.; visualization, R.C.; supervision, R.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was granted by project PDI-PFE-CDI 2021, entitled Increasing the Performance of Scientific Research, Supporting Excellence in Medical Research and Innovation, PROGRES, no. 40PFE/30.12.2021 of Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xuan, Z.; Zachar, V.; Pennisi, C.P. Sources, Selection, and Microenvironmental Preconditioning of Cells for Urethral Tissue Engineering. Int. J. Mol. Sci. 2022, 23, 4074. [Google Scholar] [CrossRef] [PubMed]
  2. Hickling, D.R.; Sun, T.-T.; Wu, X.-R. Anatomy and Physiology of the Urinary Tract: Relation to Host Defense and Microbial Infection. Microbiol. Spectr. 2015, 3, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Coorevits, L.; Heytens, S.; Boelens, J.; Claeys, G. The resident microflora of voided midstream urine of healthy controls: Standard versus expanded urine culture protocols. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 635–639. [Google Scholar] [CrossRef] [PubMed]
  4. Hilt, E.E.; McKinley, K.; Pearce, M.M.; Rosenfeld, A.B.; Zilliox, M.J.; Mueller, E.R.; Brubaker, L.; Gai, X.; Wolfe, A.J.; Schreckenberger, P.C. Urine is not sterile: Use of enhanced urine culture techniques to detect resident bacterial flora in the adult female bladder. J. Clin. Microbiol. 2014, 52, 871–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rajas, O.; Quirós, L.M.; Ortega, M.; Vazquez-Espinosa, E.; Merayo-Lloves, J.; Vazquez, F.; García, B. Glycosaminoglycans are involved in bacterial adherence to lung cells. BMC Infect. Dis. 2017, 17, 319. [Google Scholar] [CrossRef] [Green Version]
  6. Rose, C.; Parker, A.; Jefferson, B.; Cartmell, E. The Characterization of Feces and Urine: A Review of the Literature to Inform Advanced Treatment Technology. Crit. Rev. Environ. Sci. Technol. 2015, 45, 1827. [Google Scholar] [CrossRef] [Green Version]
  7. Iwakiri, J.; Freiha, F.S.; Shortliffe, L.M. Prospective study of urinary tract infections and urinary antibodies after radical prostatectomy and cystoprostatectomy. Urol. Clin. N. Am. 2002, 29, 251–258. [Google Scholar] [CrossRef]
  8. Zeimet, A.; McBride, D.R.; Basilan, R.; Roland, W.E.; McCrary, D.; Hoonmo, K. Infectious Diseases. Textb. Fam. Med. 2011, 207, 1160. [Google Scholar] [CrossRef]
  9. Chiu, L.; Bazin, T.; Truchetet, M.E.; Schaeverbeke, T.; Delhaes, L.; Pradeu, T. Protective Microbiota: From Localized to Long-Reaching Co-Immunity. Front. Immunol. 2017, 8, 1678. [Google Scholar] [CrossRef]
  10. Warzecha, D.; Pietrzak, B.; Urban, A.; Wielgoś, M. How to avoid drug resistance during treatment and prevention of urinary tract infections. Prz. Menopauzalny 2021, 20, 111715. [Google Scholar] [CrossRef]
  11. Foxman, B. The epidemiology of urinary tract infection. Nat. Rev. Urol. 2010, 7, 190. [Google Scholar] [CrossRef]
  12. Chenoweth, C.; Saint, S. Preventing Catheter-Associated Urinary Tract Infections in the Intensive Care Unit. Crit. Care Clin. 2013, 29, 5. [Google Scholar] [CrossRef] [PubMed]
  13. Simmering, J.E.; Tang, F.; Cavanaugh, J.E.; Polgreen, L.A.; Polgreen, P.M. The Increase in Hospitalizations for Urinary Tract Infections and the Associated Costs in the United States, 1998–2011. Open Forum Infect. Dis. 2017, 4, 281. [Google Scholar] [CrossRef] [PubMed]
  14. Historical|CMS. Available online: https://www.cms.gov/Research-Statistics-Data-and-Systems/Statistics-Trends-and-Reports/NationalHealthExpendData/NationalHealthAccountsHistorical (accessed on 20 January 2023).
  15. François, M.; Hanslik, T.; Dervaux, B.; Le Strat, Y.; Souty, C.; Vaux, S.; Maugat, S.; Rondet, C.; Sarazin, M.; Heym, B.; et al. The economic burden of urinary tract infections in women visiting general practices in France: A cross-sectional survey. BMC Health Serv. Res. 2016, 16, 365. [Google Scholar] [CrossRef] [PubMed]
  16. Iskandar, K.; Rizk, R.; Matta, R.; Husni-Samaha, R.; Sacre, H.; Bouraad, E.; Dirani, N.; Salameh, P.; Molinier, L.; Roques, C.; et al. Economic Burden of Urinary Tract Infections From Antibiotic-Resistant Escherichia coli Among Hospitalized Adult Patients in Lebanon: A Prospective Cohort Study. Value Health Reg. Issues 2021, 24, 6. [Google Scholar] [CrossRef]
  17. Thorpe, K.E.; Joski, P.; Johnston, K.J. Antibiotic-resistant infection treatment costs have doubled since 2002, now exceeding $2 billion annually. Health Aff. 2018, 37, 1153. [Google Scholar] [CrossRef]
  18. Najar, M.S.; Saldanha, C.L.; Banday, K.A. Approach to urinary tract infections. Indian J. Nephrol. 2009, 19, 129–139. [Google Scholar] [CrossRef] [PubMed]
  19. Herberg, J.; Pahari, A.; Walters, S.; Levin, M. Infectious Diseases and the Kidney. Pediatr. Nephrol. 2009, 1235–1273. [Google Scholar] [CrossRef]
  20. Kantele, A.; Palkola, N.; Arvilommi, H.; Honkinen, O.; Jahnukainen, T.; Mertsola, J.; Kantele, J.M. Local Immune Response to Upper Urinary Tract Infections in Children. Clin. Vaccine Immunol. 2008, 15, 412. [Google Scholar] [CrossRef] [Green Version]
  21. Guclu, E.; Halis, F.; Kose, E.; Ogutlu, A.; Karabay, O. Risk factors of multidrug-resistant bacteria in community-acquired urinary tract infections. Afr. Health Sci. 2021, 21, 214. [Google Scholar] [CrossRef]
  22. El Chakhtoura, N.G.; Bonomo, R.A.; Jump, R.L.P. Influence of Aging and Environment on Presentation of Infection in Older Adults. Infect. Dis. Clin. N. Am. 2017, 31, 593. [Google Scholar] [CrossRef] [PubMed]
  23. Gajdács, M.; Ábrók, M.; Lázár, A.; Burián, K. Urinary Tract Infections in Elderly Patients: A 10-Year Study on Their Epidemiology and Antibiotic Resistance Based on the WHO Access, Watch, Reserve (AWaRe) Classification. Antibiotocs 2021, 10, 1098. [Google Scholar] [CrossRef] [PubMed]
  24. Abraham, S.N.; Miao, Y. The nature of immune responses to urinary tract infections. Nat. Rev. Immunol. 2015, 15, 655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Spencer, J.D.; Schwaderer, A.L.; Becknell, B.; Watson, J.; Hains, D.S. The innate immune response during urinary tract infection and pyelonephritis. Pediatr. Nephrol. 2014, 29, 1139. [Google Scholar] [CrossRef] [Green Version]
  26. Cedzyński, M.; Thielens, N.M.; Mollnes, T.E.; Vorup-Jensen, T. Editorial: The Role of Complement in Health and Disease. Front. Immunol. 2019, 10, 1869. [Google Scholar] [CrossRef] [PubMed]
  27. Abreu, A.G.; Barbosa, A.S. How Escherichia coli Circumvent Complement-Mediated Killing. Front. Immunol. 2017, 8, 452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Zandbergen, L.E.; Halverson, T.; Brons, J.K.; Wolfe, A.J.; Vos, M.G.J. de The Good and the Bad: Ecological Interaction Measurements Between the Urinary Microbiota and Uropathogens. Front. Microbiol. 2021, 12, 659450. [Google Scholar] [CrossRef]
  29. Zagaglia, C.; Ammendolia, M.G.; Maurizi, L.; Nicoletti, M.; Longhi, C. Urinary Tract Infections Caused by Uropathogenic Escherichia coli Strains—New Strategies for an Old Pathogen. Microorganisms 2022, 10, 1425. [Google Scholar] [CrossRef]
  30. Marrs, C.F.; Zhang, L.; Foxman, B. Escherichia coli mediated urinary tract infections: Are there distinct uropathogenic E. coli (UPEC) pathotypes? FEMS Microbiol. Lett. 2005, 252, 183–190. [Google Scholar] [CrossRef] [Green Version]
  31. Terlizzi, M.E.; Gribaudo, G.; Maffei, M.E. UroPathogenic Escherichia coli (UPEC) infections: Virulence factors, bladder responses, antibiotic, and non-antibiotic antimicrobial strategies. Front. Microbiol. 2017, 8, 1566. [Google Scholar] [CrossRef]
  32. Kline, K.A.; Lewis, A.L. Gram-Positive Uropathogens, Polymicrobial Urinary Tract Infection, and the Emerging Microbiota of the Urinary Tract. Microbiol. Spectr. 2016, 4, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Hochstedler, B.R.; Burnett, L.; Price, T.K.; Jung, C.; Wolfe, A.J.; Brubaker, L. Urinary microbiota of women with recurrent urinary tract infection: Collection and culture methods. Int. Urogynecol. J. 2021, 33, 563–570. [Google Scholar] [CrossRef] [PubMed]
  34. Behzadi, P.; Behzadi, E.; Ranjbar, R. Urinary tract infections and Candida albicans. Cent. Eur. J. Urol. 2015, 68, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Sakhi, H.; Join-Lambert, O.; Goujon, A.; Culty, T.; Loubet, P.; Dang, J.; Drouot, S.; De Bayser, H.; Michaud, C.; Ghislain, L.; et al. Encrusted Urinary Tract Infections Due to Corynebacteria Species. Kidney Int. Reports 2021, 6, 179–186. [Google Scholar] [CrossRef]
  36. Flores-Mireles, A.L.; Walker, J.N.; Caparon, M.; Hultgren, S.J. Urinary tract infections: Epidemiology, mechanisms of infection and treatment options. Nat. Rev. Microbiol. 2015, 13, 269–284. [Google Scholar] [CrossRef]
  37. Echavarría, M. Adenoviruses in Immunocompromised Hosts. Clin. Microbiol. Rev. 2008, 21, 704. [Google Scholar] [CrossRef] [Green Version]
  38. Al-Hilu, S.A.; Al-Shujairi, W.H. Dual Role of Bacteria in Carcinoma: Stimulation and Inhibition. Int. J. Microbiol. 2020, 2020, 4639761. [Google Scholar] [CrossRef]
  39. Bernardes, N.; Seruca, R.; Chakrabarty, A.M.; Fialho, A.M. Microbial-based therapy of Cancer Current progress and future prospects. Bioeng. Bugs 2010, 1, 178–190. [Google Scholar] [CrossRef] [Green Version]
  40. Łukasiewicz, K.; Fol, M. Microorganisms in the Treatment of Cancer: Advantages and Limitations. J. Immunol. Res. 2018, 2018, 2397808. [Google Scholar] [CrossRef] [Green Version]
  41. Dalhoff, A. Selective toxicity of antibacterial agents—Still a valid concept or do we miss chances and ignore risks? Infection 2021, 49, 29–56. [Google Scholar] [CrossRef]
  42. Egorov, A.M.; Ulyashova, M.M.; Rubtsova, M.Y. Bacterial enzymes and antibiotic resistance. Acta Naturae 2018, 10, 33–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Pham, J.V.; Yilma, M.A.; Feliz, A.; Majid, M.T.; Maffetone, N.; Walker, J.R.; Kim, E.; Cho, H.J.; Reynolds, J.M.; Song, M.C.; et al. A review of the microbial production of bioactive natural products and biologics. Front. Microbiol. 2019, 10, 1404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Purssell, E. Antimicrobials. In Understanding Pharmacology in Nursing Practice; Springer International Publishing: Cham, Switzerland, 2020; pp. 147–165. [Google Scholar] [CrossRef]
  45. Tan, S.Y.; Tatsumura, Y. Alexander Fleming (1881–1955): Discoverer of penicillin. Singapore Med. J. 2015, 56, 366–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Aminov, R.I. A brief history of the antibiotic era: Lessons learned and challenges for the future. Front. Microbiol. 2010, 1, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Buchholz, K. A breakthrough in enzyme technology to fight penicillin resistance—Industrial application of penicillin amidase. Appl. Microbiol. Biotechnol. 2016, 100, 3825–3839. [Google Scholar] [CrossRef]
  48. Podolsky, S.H. The evolving response to antibiotic resistance (1945–2018). Palgrave Commun. 2018, 4, 124. [Google Scholar] [CrossRef] [Green Version]
  49. Ventola, C.L. The antibiotic resistance crisis: Part 1: Causes and threats. Pharm. Ther. 2015, 40, 277–283. [Google Scholar]
  50. Piddock, L.J.V.; Levy, S.; Marshall, B.; Society, R.; Spellberg, B.; Blaser, M.; Guidos, R.; Cars, O.; Hedin, A. The crisis of no new antibiotics--what is the way forward? Lancet. Infect. Dis. 2012, 12, 249–253. [Google Scholar] [CrossRef]
  51. Kohanski, M.A.; Dwyer, D.J.; Collins, J.J. How antibiotics kill bacteria: From targets to networks. Nat. Rev. Microbiol. 2010, 8, 423–435. [Google Scholar] [CrossRef] [Green Version]
  52. Stokes, J.M.; Lopatkin, A.J.; Lobritz, M.A.; Collins, J.J. Bacterial Metabolism and Antibiotic Efficacy. Cell Metab. 2019, 30, 251–259. [Google Scholar] [CrossRef]
  53. Lade, H.; Kim, J.-S. Bacterial Targets of Antibiotics in Methicillin-Resistant Staphylococcus aureus. Antibiotics 2021, 10, 398. [Google Scholar] [CrossRef] [PubMed]
  54. Soares, G.M.S.; Figueiredo, L.C.; Faveri, M.; Cortelli, S.C.; Duarte, P.M.; Feres, M. Mechanisms of action of systemic antibiotics used in periodontal treatment and mechanisms of bacterial resistance to these drugs. J. Appl. Oral Sci. 2012, 20, 295–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schenone, M.; Dančík, V.; Wagner, B.K.; Clemons, P.A. Target identification and mechanism of action in chemical biology and drug discovery. Nat. Chem. Biol. 2013, 9, 232–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Song, H.P.; Wu, S.Q.; Hao, H.; Chen, J.; Lu, J.; Xu, X.; Li, P.; Yang, H. A chemical family-based strategy for uncovering hidden bioactive molecules and multicomponent interactions in herbal medicines. Sci. Rep. 2016, 6, 23840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Frieri, M.; Kumar, K.; Boutin, A. Antibiotic resistance. J. Infect. Public Health 2017, 10, 369–378. [Google Scholar] [CrossRef] [Green Version]
  58. Prestinaci, F.; Pezzotti, P.; Pantosti, A. Antimicrobial resistance: A global multifaceted phenomenon. Pathog. Glob. Health 2015, 109, 309–318. [Google Scholar] [CrossRef] [Green Version]
  59. Davies, J.; Davies, D. Origins and evolution of antibiotic resistance. Microbiol. Mol. Biol. Rev. 2010, 74, 417–433. [Google Scholar] [CrossRef] [Green Version]
  60. Aghapour, Z.; Gholizadeh, P.; Ganbarov, K.; Bialvaei, A.Z.; Mahmood, S.S.; Tanomand, A.; Yousefi, M.; Asgharzadeh, M.; Yousefi, B.; Kafil, H.S. Molecular mechanisms related to colistin resistance in enterobacteriaceae. Infect. Drug Resist. 2019, 12, 965–975. [Google Scholar] [CrossRef] [Green Version]
  61. Codjoe, F.; Donkor, E. Carbapenem Resistance: A Review. Med. Sci. 2017, 6, 1. [Google Scholar] [CrossRef] [Green Version]
  62. Llor, C.; Bjerrum, L. Antimicrobial resistance: Risk associated with antibiotic overuse and initiatives to reduce the problem. Ther. Adv. Drug Saf. 2014, 5, 229–241. [Google Scholar] [CrossRef]
  63. van Belkum, A.; Bachmann, T.T.; Lüdke, G.; Lisby, J.G.; Kahlmeter, G.; Mohess, A.; Becker, K.; Hays, J.P.; Woodford, N.; Mitsakakis, K.; et al. Developmental roadmap for antimicrobial susceptibility testing systems. Nat. Rev. Microbiol. 2019, 17, 51–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Tenover, F.C. Mechanisms of antimicrobial resistance in bacteria. Am. J. Infect. Control 2006, 34, S3–S10. [Google Scholar] [CrossRef] [PubMed]
  65. de Lima Procópio, R.E.; da Silva, I.R.; Martins, M.K.; de Azevedo, J.L.; de Araújo, J.M. Antibiotics produced by Streptomyces. Brazilian J. Infect. Dis. 2012, 16, 466–471. [Google Scholar] [CrossRef] [Green Version]
  66. Hoffman, S.B. Mechanisms of Antibiotic Resistance. Compend. Contin. Educ. Pract. Vet. 2001, 23, 464–472. [Google Scholar] [CrossRef] [Green Version]
  67. Peterson, E.; Kaur, P. Antibiotic resistance mechanisms in bacteria: Relationships between resistance determinants of antibiotic producers, environmental bacteria, and clinical pathogens. Front Microbiol. 2018, 9, 2928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Bakkeren, E.; Diard, M.; Hardt, W.D. Evolutionary causes and consequences of bacterial antibiotic persistence. Nat. Rev. Microbiol. 2020, 18, 479–490. [Google Scholar] [CrossRef] [PubMed]
  69. Martínez, J.L.; Baquero, F. Interactions among strategies associated with bacterial infection: Pathogenicity, epidemicity, and antibiotic resistance. Clin. Microbiol. Rev. 2002, 15, 647–679. [Google Scholar] [CrossRef] [Green Version]
  70. Beceiro, A.; Tomás, M.; Bou, G. Antimicrobial resistance and virulence: A successful or deleterious association in the bacterial world? Clin. Microbiol. Rev. 2013, 26, 185–230. [Google Scholar] [CrossRef] [Green Version]
  71. Corona, F.; Martinez, J. Phenotypic Resistance to Antibiotics. Antibiotics 2013, 2, 237–255. [Google Scholar] [CrossRef] [Green Version]
  72. Li, X.Z.; Nikaido, H. Efflux-mediated drug resistance in bacteria: An update. Drugs 2009, 69, 1555–1623. [Google Scholar] [CrossRef]
  73. Reygaert, C.W. An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 2018, 4, 482–501. [Google Scholar] [CrossRef] [PubMed]
  74. Delcour, A.H. Outer membrane permeability and antibiotic resistance. Biochim. Biophys. Acta—Proteins Proteomics 2009, 1794, 808–816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Blair, J.M.A.; Webber, M.A.; Baylay, A.J.; Ogbolu, D.O.; Piddock, L.J.V. Molecular mechanisms of antibiotic resistance. Nat. Rev. Microbiol. 2015, 13, 42–51. [Google Scholar] [CrossRef] [PubMed]
  76. Miller, S.I. Antibiotic Resistance and Regulation of the Gram-Negative Bacterial Outer Membrane Barrier by Host Innate Immune Molecules. MBio 2016, 7, 16. [Google Scholar] [CrossRef] [Green Version]
  77. Okuda, S.; Sherman, D.J.; Silhavy, T.J.; Ruiz, N.; Kahne, D. Lipopolysaccharide transport and assembly at the outer membrane: The PEZ model. Nat. Rev. Microbiol. 2016, 14, 337–345. [Google Scholar] [CrossRef] [Green Version]
  78. Lehman, K.M.; Grabowicz, M. Countering gram-negative antibiotic resistance: Recent progress in disrupting the outer membrane with novel therapeutics. Antibiotics 2019, 8, 163. [Google Scholar] [CrossRef] [Green Version]
  79. Poirel, L.; Jayol, A.; Nordmanna, P. Polymyxins: Antibacterial activity, susceptibility testing, and resistance mechanisms encoded by plasmids or chromosomes. Clin. Microbiol. Rev. 2017, 30, 557–596. [Google Scholar] [CrossRef] [Green Version]
  80. Falagas, M.E.; Rafailidis, P.I.; Matthaiou, D.K. Resistance to polymyxins: Mechanisms, frequency and treatment options. Drug Resist. Updat. 2010, 13, 132–138. [Google Scholar] [CrossRef]
  81. El-Sayed Ahmed, M.A.E.G.; Zhong, L.L.; Shen, C.; Yang, Y.; Doi, Y.; Tian, G.B. Colistin and its role in the Era of antibiotic resistance: An extended review (2000–2019). Emerg. Microbes Infect. 2020, 9, 868–885. [Google Scholar] [CrossRef] [Green Version]
  82. Alekshun, M.N.; Levy, S.B. Molecular Mechanisms of Antibacterial Multidrug Resistance. Cell 2007, 128, 1037–1050. [Google Scholar] [CrossRef]
  83. Pang, Z.; Raudonis, R.; Glick, B.R.; Lin, T.J.; Cheng, Z. Antibiotic resistance in Pseudomonas aeruginosa: Mechanisms and alternative therapeutic strategies. Biotechnol. Adv. 2019, 37, 177–192. [Google Scholar] [CrossRef] [PubMed]
  84. Poole, K. Efflux pumps as antimicrobial resistance mechanisms. Ann Med. 2007, 39, 162–176. [Google Scholar] [CrossRef] [PubMed]
  85. De Kraker, M.E.A.; Stewardson, A.J.; Harbarth, S. Will 10 Million People Die a Year due to Antimicrobial Resistance by 2050? PLoS Med. 2016, 13, 1002184. [Google Scholar] [CrossRef] [Green Version]
  86. Paul, R. State of the globe: Rising antimicrobial resistance of pathogens in urinary tract infection. J. Glob. Infect. Dis. 2018, 10, 117–118. [Google Scholar] [CrossRef]
  87. Reza Mortazavi-Tabatabaei, S.; Ghaderkhani, J.; Nazari, A.; Sayehmiri, K.; Sayehmiri, F.; Pakzad, I. Pattern of antibacterial resistance in urinary tract infections: A systematic review and meta-analysis. Int. J. Prev. Med. 2019, 10, 169. [Google Scholar] [CrossRef]
  88. Doron, S.; Gorbach, S.L. Bacterial infections: Overview. In International Encyclopedia of Public Health; Elsevier: Amsterdam, The Netherlands, 2008; pp. 273–282. ISBN 9780123739605. [Google Scholar]
  89. Arvanitis, M.; Anagnostou, T.; Fuchs, B.B.; Caliendo, A.M.; Mylonakis, E. Molecular and nonmolecular diagnostic methods for invasive fungal infections. Clin. Microbiol. Rev. 2014, 27, 490–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. El-Sayed, A.; Kamel, M. Advances in nanomedical applications: Diagnostic, therapeutic, immunization, and vaccine production. Environ. Sci. Pollut. Res. 2020, 27, 19200–19213. [Google Scholar] [CrossRef]
  91. Kaittanis, C.; Santra, S.; Perez, J.M. Emerging nanotechnology-based strategies for the identification of microbial pathogenesis. Adv. Drug Deliv. Rev. 2010, 62, 408–423. [Google Scholar] [CrossRef] [Green Version]
  92. Zhu, X.; Radovic-Moreno, A.F.; Wu, J.; Langer, R.; Shi, J. Nanomedicine in the management of microbial infection—Overview and perspectives. Nano Today 2014, 9, 478–498. [Google Scholar] [CrossRef] [Green Version]
  93. Buzea, C.; Pacheco, I.I.; Robbie, K. Nanomaterials and nanoparticles: Sources and toxicity. Biointerphases 2007, 2, MR17–MR71. [Google Scholar] [CrossRef]
  94. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
  95. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; 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 10 Technology 1007 Nanotechnology 03 Chemical Sciences 0306 Physical Chemistry (incl. Structural) 03 Chemical Sciences 0303 Macromolecular and Materials Chemistry 11 Medical and Health Sciences 1115 Pharmacology and Pharmaceutical Sciences 09 Engineering 0903 Biomedical Engineering Prof Ueli Aebi, Prof Peter Gehr. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [Green Version]
  96. Li, H.; Yang, Y.; Hong, W.; Huang, M.; Wu, M.; Zhao, X. Applications of genome editing technology in the targeted therapy of human diseases: Mechanisms, advances and prospects. Signal Transduct. Target. Ther. 2020, 5, 1–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Li, B.; Webster, T.J. Bacteria antibiotic resistance: New challenges and opportunities for implant-associated orthopedic infections. J. Orthop. Res. 2018, 36, 22–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Mambatta, A.; Rashme, V.; Menon, S.; Jayarajan, J.; Harini, S.; Kuppusamy, J. Reliability of dipstick assay in predicting urinary tract infection. J. Fam. Med. Prim. Care 2015, 4, 265. [Google Scholar] [CrossRef]
  99. Álvarez-Barrientos, A.; Arroyo, J.; Cantón, R.; Nombela, C.; Sánchez-Pérez, M. Applications of flow cytometry to clinical microbiology. Clin. Microbiol. Rev. 2000, 13, 167–195. [Google Scholar] [CrossRef] [Green Version]
  100. Davenport, M.; Mach, K.E.; Shortliffe, L.M.D.; Banaei, N.; Wang, T.H.; Liao, J.C. New and developing diagnostic technologies for urinary tract infections. Nat. Rev. Urol. 2017, 14, 298–310. [Google Scholar] [CrossRef] [Green Version]
  101. Murray, P.R. The Clinician and the Microbiology Laboratory. In Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases; Elsevier: Amsterdam, The Netherlands, 2014; Volume 1, pp. 191–223. ISBN 9996096742. [Google Scholar]
  102. Bhalla, N.; Pan, Y.; Yang, Z.; Payam, A.F. Opportunities and Challenges for Biosensors and Nanoscale Analytical Tools for Pandemics: COVID-19. ACS Nano 2020, 14, 7783–7807. [Google Scholar] [CrossRef]
  103. Wei, F.; Lillehoj, P.B.; Ho, C.M. DNA diagnostics: Nanotechnology-enhanced electrochemical detection of nucleic acids. Pediatr. Res. 2010, 67, 458–468. [Google Scholar] [CrossRef] [Green Version]
  104. Bhalla, N.; Jolly, P.; Formisano, N.; Estrela, P. Introduction to biosensors. Essays Biochem. 2016, 60, 1–8. [Google Scholar] [CrossRef]
  105. Nguyen, H.H.; Lee, S.H.; Lee, U.J.; Fermin, C.D.; Kim, M. Immobilized enzymes in biosensor applications. Materials 2019, 12, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Hasan, A.; Nurunnabi, M.; Morshed, M.; Paul, A.; Polini, A.; Kuila, T.; Al Hariri, M.; Lee, Y.K.; Jaffa, A.A. Recent advances in application of biosensors in tissue engineering. Biomed Res. Int. 2014, 2014, 307519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Mehrotra, P. Biosensors and their applications—A review. J. Oral Biol. Craniofacial Res. 2016, 6, 153–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Mach, K.E.; Wong, P.K.; Liao, J.C. Biosensor diagnosis of urinary tract infections: A path to better treatment? Trends Pharmacol. Sci. 2011, 32, 330–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Roca, I.; Akova, M.; Baquero, F.; Carlet, J.; Cavaleri, M.; Coenen, S.; Cohen, J.; Findlay, D.; Gyssens, I.; Heure, O.E.; et al. The global threat of antimicrobial resistance: Science for intervention. New Microbes New Infect. 2015, 6, 22–29. [Google Scholar] [CrossRef] [Green Version]
  110. Fair, R.J.; Tor, Y. Antibiotics and bacterial resistance in the 21st century. Perspect. Medicin. Chem. 2014, 6, 25–64. [Google Scholar] [CrossRef] [Green Version]
  111. Wistrand-Yuen, E.; Knopp, M.; Hjort, K.; Koskiniemi, S.; Berg, O.G.; Andersson, D.I. Evolution of high-level resistance during low-level antibiotic exposure. Nat. Commun. 2018, 9, 1599. [Google Scholar] [CrossRef] [Green Version]
  112. Kapoor, G.; Saigal, S.; Elongavan, A. Action and resistance mechanisms of antibiotics: A guide for clinicians. J. Anaesthesiol. Clin. Pharmacol. 2017, 33, 300–305. [Google Scholar] [CrossRef]
  113. Alanis, A.J. Resistance to antibiotics: Are we in the post-antibiotic era? Arch. Med. Res. 2005, 36, 697–705. [Google Scholar] [CrossRef]
  114. Ray, S.; Das, S.; Suar, M. Molecular mechanism of drug resistance. In Drug Resistance in Bacteria, Fungi, Malaria, and Cancer; Springer International Publishing: New York, NY, USA, 2017; pp. 47–110. ISBN 9783319486833. [Google Scholar]
  115. Abo-zeid, Y.; Williams, G.R. The potential anti-infective applications of metal oxide nanoparticles: A systematic review. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, 1592. [Google Scholar] [CrossRef]
  116. Eleraky, N.E.; Allam, A.; Hassan, S.B.; Omar, M.M. Nanomedicine fight against antibacterial resistance: An overview of the recent pharmaceutical innovations. Pharmaceutics 2020, 12, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Yeh, Y.C.; Huang, T.H.; Yang, S.C.; Chen, C.C.; Fang, J.Y. Nano-Based Drug Delivery or Targeting to Eradicate Bacteria for Infection Mitigation: A Review of Recent Advances. Front. Chem. 2020, 8, 286. [Google Scholar] [CrossRef] [Green Version]
  118. Ruddaraju, L.K.; Pammi, S.V.N.; Guntuku, G.S.; Padavala, V.S.; Kolapalli, V.R.M. A review on anti-bacterials to combat resistance: From ancient era of plants and metals to present and future perspectives of green nano technological combinations. Asian J. Pharm. Sci. 2020, 15, 42–59. [Google Scholar] [CrossRef] [PubMed]
  119. Thapa, R.K.; Diep, D.B.; Tønnesen, H.H. Nanomedicine-based antimicrobial peptide delivery for bacterial infections: Recent advances and future prospects. J. Pharm. Investig. 2021, 51, 377–398. [Google Scholar] [CrossRef]
  120. Wang, L.; Hu, C.; Shao, L. The antimicrobial activity of nanoparticles: Present situation and prospects for the future. Int. J. Nanomed. 2017, 12, 1227–1249. [Google Scholar] [CrossRef] [Green Version]
  121. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2020, 20, 101–124. [Google Scholar] [CrossRef]
  122. Demetzos, C. Application of Nanotechnology in Drug Delivery and Targeting. Pharm. Nanotechnol. 2016, 77–145. [Google Scholar] [CrossRef]
  123. Djeribi, R.; Bouchloukh, W.; Jouenne, T.; Menaa, B. Characterization of bacterial biofilms formed on urinary catheters. Am. J. Infect. Control 2012, 40, 854–859. [Google Scholar] [CrossRef]
  124. Jamal, M.; Ahmad, W.; Andleeb, S.; Jalil, F.; Imran, M.; Nawaz, M.A.; Hussain, T.; Ali, M.; Rafiq, M.; Kamil, M.A. Bacterial biofilm and associated infections. J. Chin. Med. Assoc. 2018, 81, 7–11. [Google Scholar] [CrossRef]
  125. Anh, N.H.; Kim, S.J.; Long, N.P.; Min, J.E.; Yoon, Y.C.; Lee, E.G.; Kim, M.; Kim, T.J.; Yang, Y.Y.; Son, E.Y.; et al. Ginger on Human Health: A Comprehensive Systematic Review of 109 Randomized Controlled Trials. Nutrient 2020, 12, 157. [Google Scholar] [CrossRef]
  126. Chadha, J.; Harjai, K.; Chhibber, S. Repurposing phytochemicals as anti-virulent agents to attenuate quorum sensing-regulated virulence factors and biofilm formation in Pseudomonas aeruginosa. Microb. Biotechnol. 2022, 15, 1695. [Google Scholar] [CrossRef] [PubMed]
  127. Vadekeetil, A.; Chhibber, S.; Harjai, K. Efficacy of intravesical targeting of novel quorum sensing inhibitor nanoparticles against Pseudomonas aeruginosa biofilm-associated murine pyelonephritis. J. Drug Target. 2019, 27, 995–1003. [Google Scholar] [CrossRef]
  128. Sharma, K.; Bose, S.K.; Chhibber, S.; Harjai, K. Exploring the Therapeutic Efficacy of Zingerone Nanoparticles in Treating Biofilm-Associated Pyelonephritis Caused by Pseudomonas aeruginosa in the Murine Model. Inflammation 2020, 43, 2344–2356. [Google Scholar] [CrossRef] [PubMed]
  129. Brauner, B.; Semmler, J.; Rauch, D.; Nokaj, M.; Haiss, P.; Schwarz, P.; Wirth, M.; Gabor, F. Trimethoprim-Loaded PLGA NanoparticlesGrafted with WGA as Potential Intravesical Therapy of Urinary TractInfections—Studies on Adhesion to SV-HUCs Under Varying Time, pH, and Drug-Loading Conditions. ACS Omega 2020, 5, 17377. [Google Scholar] [CrossRef] [PubMed]
  130. 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]
  131. Sánchez, S.V.; Navarro, N.; Catalán-Figueroa, J.; Morales, J.O. Nanoparticles as Potential Novel Therapies for Urinary Tract Infections. Front. Cell. Infect. Microbiol. 2021, 11, 292. [Google Scholar] [CrossRef]
  132. Gasión, J.P.B.; Cruz, J.F.J. Improving efficacy of intravesical chemotherapy. Eur. Urol. 2006, 50, 225–234. [Google Scholar] [CrossRef] [PubMed]
  133. Pichl, C.M.; Austria, B.D.; Brauner, B.; Gabor, F.; Wirth, M.; Neutsch, L. Biomimickry of UPEC Cytoinvasion: A Novel Concept for Improved Drug Delivery in UTI. Pathog. 2016, 5, 16. [Google Scholar] [CrossRef] [Green Version]
  134. Alsaab, H.O.; Alharbi, F.D.; Alhibs, A.S.; Alanazi, N.B.; Alshehri, B.Y.; Saleh, M.A.; Alshehri, F.S.; Algarni, M.A.; Almugaiteeb, T.; Uddin, M.N.; et al. PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases. Pharmaceutics 2022, 14, 2728. [Google Scholar] [CrossRef]
  135. Breijyeh, Z.; Jubeh, B.; Karaman, R. Resistance of Gram-Negative Bacteria to Current Antibacterial Agents and Approaches to Resolve It. Molecules 2020, 25, 1340. [Google Scholar] [CrossRef]
  136. Uddin, T.M.; Chakraborty, A.J.; Khusro, A.; Zidan, B.R.M.; Mitra, S.; Bin Emran, T.; Dhama, K.; Ripon, M.K.H.; Gajdács, M.; Sahibzada, M.U.K.; et al. Antibiotic resistance in microbes: History, mechanisms, therapeutic strategies and future prospects. J. Infect. Public Health 2021, 14, 1750–1766. [Google Scholar] [CrossRef] [PubMed]
  137. Lee, N.Y.; Ko, W.C.; Hsueh, P.R. Nanoparticles in the treatment of infections caused by multidrug-resistant organisms. Front. Pharmacol. 2019, 10, 1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Ferreira, M.; Ogren, M.; Dias, J.N.R.; Silva, M.; Gil, S.; Tavares, L.; Aires-Da-silva, F.; Gaspar, M.M.; Aguiar, S.I. Liposomes as Antibiotic Delivery Systems: A Promising Nanotechnological Strategy against Antimicrobial Resistance. Molecules 2021, 26, 2047. [Google Scholar] [CrossRef]
  139. Mugabe, C.; Halwani, M.; Azghani, A.O.; Lafrenie, R.M.; Omri, A. Mechanism of Enhanced Activity of Liposome-Entrapped Aminoglycosides against Resistant Strains of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2006, 50, 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Maxwell, A.; Ghate, V.; Aranjani, J.; Lewis, S. Breaking the barriers for the delivery of amikacin: Challenges, strategies, and opportunities. Life Sci. 2021, 284, 119883. [Google Scholar] [CrossRef] [PubMed]
  141. Sartaj, A.; Qamar, Z.; Qizilbash, F.F.; Annu; Md, S.; Alhakamy, N.A.; Baboota, S.; Ali, J. Polymeric Nanoparticles: Exploring the Current Drug Development and Therapeutic Insight of Breast Cancer Treatment and Recommendations. Polymers 2021, 13, 4400. [Google Scholar] [CrossRef] [PubMed]
  142. Song, J.; Kook, M.S.; Kim, B.H.; Jeong, Y.I.; Oh, K.J. Ciprofloxacin-releasing ROS-Sensitive nanoparticles composed of poly(Ethylene Glycol)/poly(D,L-lactide-coglycolide) for antibacterial treatment. Materials 2021, 14, 4125. [Google Scholar] [CrossRef]
  143. de Llano, D.G.; Moreno-Arribas, M.V.; Bartolomé, B. Cranberry Polyphenols and Prevention against Urinary Tract Infections: Relevant Considerations. Molecules 2020, 25, 3523. [Google Scholar] [CrossRef]
  144. Urena-Saborio, H.; Udayan, A.P.M.; Alfaro-Viquez, E.; Madrigal-Carballo, S.; Reed, J.D.; Gunasekaran, S. Cranberry Proanthocyanidins-PANI Nanocomposite for the Detection of Bacteria Associated with Urinary Tract Infections. Biosensors 2021, 11, 199. [Google Scholar] [CrossRef]
  145. Hisano, M.; Bruschini, H.; Nicodemo, A.C.; Srougi, M. Cranberries and lower urinary tract infection prevention. Clinics 2012, 67, 661–667. [Google Scholar] [CrossRef]
  146. Charpentier, M.; Gutierrez, C.; Guillaudeux, T.; Verhoest, G.; Pedeux, R. Noninvasive urine-based tests to diagnose or detect recurrence of bladder cancer. Cancers 2021, 13, 1650. [Google Scholar] [CrossRef] [PubMed]
  147. Steinberg, H.E.; Bowman, N.M.; Diestra, A.; Ferradas, C.; Russo, P.; Clark, D.E.; Zhu, D.; Magni, R.; Malaga, E.; Diaz, M.; et al. Detection of toxoplasmic encephalitis in HIV positive patients in urine with hydrogel nanoparticles. PLoS Negl. Trop. Dis. 2021, 15, e0009199. [Google Scholar] [CrossRef] [PubMed]
  148. Lee, S.-B.; Lee, T.-G. Toxoplasmic Encephalitis in Patient with Acquired Immunodeficiency Syndrome. Brain Tumor Res. Treat. 2017, 5, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. El-Sayyad, G.S.; El-Bastawisy, H.S.; Gobara, M.; El-Batal, A.I. Gentamicin-Assisted Mycogenic Selenium Nanoparticles Synthesized Under Gamma Irradiation for Robust Reluctance of Resistant Urinary Tract Infection-Causing Pathogens. Biol. Trace Elem. Res. 2020, 195, 323–342. [Google Scholar] [CrossRef] [PubMed]
  150. Huang, T.; Kumari, S.; Herold, H.; Bargel, H.; Aigner, T.B.; Heath, D.E.; O’brien-Simpson, N.M.; O’connor, A.J.; Scheibel, T. Enhanced Antibacterial Activity of Se Nanoparticles Upon Coating with Recombinant Spider Silk Protein eADF4(κ16). Int. J. Nanomed. 2020, 15, 4275. [Google Scholar] [CrossRef] [PubMed]
  151. Cremonini, E.; Zonaro, E.; Donini, M.; Lampis, S.; Boaretti, M.; Dusi, S.; Melotti, P.; Lleo, M.M.; Vallini, G. Biogenic selenium nanoparticles: Characterization, antimicrobial activity and effects on human dendritic cells and fibroblasts. Microb. Biotechnol. 2016, 9, 758. [Google Scholar] [CrossRef] [Green Version]
  152. Hashem, A.H.; Selim, T.A.; Alruhaili, M.H.; Selim, S.; Alkhalifah, D.H.M.; Al Jaouni, S.K.; Salem, S.S. Unveiling Antimicrobial and Insecticidal Activities of Biosynthesized Selenium Nanoparticles Using Prickly Pear Peel Waste. J. Funct. Biomater. 2022, 13, 112. [Google Scholar] [CrossRef]
  153. Filipović, N.; Ušjak, D.; Milenković, M.T.; Zheng, K.; Liverani, L.; Boccaccini, A.R.; Stevanović, M.M. Comparative Study of the Antimicrobial Activity of Selenium Nanoparticles With Different Surface Chemistry and Structure. Front. Bioeng. Biotechnol. 2021, 8, 624621. [Google Scholar] [CrossRef]
  154. Pinto, R.M.; Lopes-De-Campos, D.; Martins, M.C.L.; Van Dijck, P.; Nunes, C.; Reis, S. Impact of nanosystems in Staphylococcus aureus biofilms treatment. FEMS Microbiol. Rev. 2019, 43, 622. [Google Scholar] [CrossRef] [Green Version]
  155. Salem, S.S.; Badawy, M.S.E.M.; Al-Askar, A.A.; Arishi, A.A.; Elkady, F.M.; Hashem, A.H. Green Biosynthesis of Selenium Nanoparticles Using Orange Peel Waste: Characterization, Antibacterial and Antibiofilm Activities against Multidrug-Resistant Bacteria. Life 2022, 12, 893. [Google Scholar] [CrossRef]
  156. Hariharan, S.; Dharmaraj, S. Selenium and selenoproteins: it’s role in regulation of inflammation. Inflammopharmacology 2020, 28, 667. [Google Scholar] [CrossRef] [PubMed]
  157. Sánchez-López, E.; Gomes, D.; Esteruelas, G.; Bonilla, L.; Lopez-Machado, A.L.; Galindo, R.; Cano, A.; Espina, M.; Ettcheto, M.; Camins, A.; et al. Metal-Based Nanoparticles as Antimicrobial Agents: An Overview. Nanomaterials 2020, 10, 292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Adeniji, O.O.; Nontongana, N.; Okoh, J.C.; Okoh, A.I. The Potential of Antibiotics and Nanomaterial Combinations as Therapeutic Strategies in the Management of Multidrug-Resistant Infections: A Review. Int. J. Mol. Sci. 2022, 23, 15038. [Google Scholar] [CrossRef] [PubMed]
  159. Abd Elkodous, M.; El-Sayyad, G.S.; Abdel Maksoud, M.I.A.; Abdelrahman, I.Y.; Mosallam, F.M.; Gobara, M.; El-Batal, A.I. Fabrication of Ultra-Pure Anisotropic Zinc Oxide Nanoparticles via Simple and Cost-Effective Route: Implications for UTI and EAC Medications. Biol. Trace Elem. Res. 2020, 196, 297–317. [Google Scholar] [CrossRef]
  160. Sirelkhatim, A.; Mahmud, S.; Seeni, A.; Kaus, N.H.M.; Ann, L.C.; Bakhori, S.K.M.; Hasan, H.; Mohamad, D. Review on Zinc Oxide Nanoparticles: Antibacterial Activity and Toxicity Mechanism. Nano-Micro Lett. 2015, 7, 219. [Google Scholar] [CrossRef] [Green Version]
  161. Li, Y.; Liao, C.; Tjong, S.C. Recent Advances in Zinc Oxide Nanostructures with Antimicrobial Activities. Int. J. Mol. Sci. 2020, 21, 8836. [Google Scholar] [CrossRef]
  162. Alsalhi, M.S.; Devanesan, S.; Atif, M.; Alqahtani, W.S.; Nicoletti, M.; Del Serrone, P. Therapeutic Potential Assessment of Green Synthesized Zinc Oxide Nanoparticles Derived from Fennel Seeds Extract. Int. J. Nanomed. 2020, 15, 8045. [Google Scholar] [CrossRef]
  163. Klink, M.J.; Laloo, N.; Taka, A.; Pakade, V.; Monapathi, M.; Modise, J. Synthesis, Characterization and Antimicrobial Activity of Zinc Oxide Nanoparticles against Selected Waterborne Bacterial and Yeast Pathogens. Molecules 2022, 27, 3532. [Google Scholar] [CrossRef]
  164. Jin, S.E.; Jin, H.E. Antimicrobial Activity of Zinc Oxide Nano/Microparticles and Their Combinations against Pathogenic Microorganisms for Biomedical Applications: From Physicochemical Characteristics to Pharmacological Aspects. Nanomaterials 2021, 11, 263. [Google Scholar] [CrossRef]
  165. da Silva, B.L.; Abuçafy, M.P.; Manaia, E.B.; Junior, J.A.O.; Chiari-Andréo, B.G.; Pietro, R.C.L.R.; Chiavacci, L.A. Relationship Between Structure And Antimicrobial Activity Of Zinc Oxide Nanoparticles: An Overview. Int. J. Nanomedicine 2019, 14, 9395. [Google Scholar] [CrossRef]
  166. Babayevska, N.; Przysiecka, Ł.; Iatsunskyi, I.; Nowaczyk, G.; Jarek, M.; Janiszewska, E.; Jurga, S. ZnO size and shape effect on antibacterial activity and cytotoxicity profile. Sci. Rep. 2022, 12, 12134. [Google Scholar] [CrossRef] [PubMed]
  167. Sayed, H.M.; Said, M.M.; Morcos, N.Y.S.; El Gawish, M.A.; Ismail, A.F.M. Antitumor and Radiosensitizing Effects of Zinc Oxide-Caffeic AcidNanoparticles against Solid Ehrlich Carcinoma in Female Mice. Integr. Cancer Ther. 2021, 20. [Google Scholar] [CrossRef]
  168. Anjum, S.; Hashim, M.; Malik, S.A.; Khan, M.; Lorenzo, J.M.; Abbasi, B.H.; Hano, C. Recent Advances in Zinc Oxide Nanoparticles (ZnO NPs) for Cancer Diagnosis, Target Drug Delivery, and Treatment. Cancers 2021, 13, 4570. [Google Scholar] [CrossRef] [PubMed]
  169. Jiang, J.; Pi, J.; Cai, J. The Advancing of Zinc Oxide Nanoparticles for Biomedical Applications. Bioinorg. Chem. Appl. 2018, 2018, 106252. [Google Scholar] [CrossRef] [PubMed]
  170. Khan, R.; Inam, M.A.; Iqbal, M.M.; Shoaib, M.; Park, D.R.; Lee, K.H.; Shin, S.; Khan, S.; Yeom, I.T. Removal of ZnO Nanoparticles from Natural Waters by Coagulation-Flocculation Process: Influence of Surfactant Type on Aggregation, Dissolution and Colloidal Stability. Sustainability 2018, 11, 17. [Google Scholar] [CrossRef] [Green Version]
  171. Vasudevan, S.; Srinivasan, P.; Rayappan, J.B.B.; Solomon, A.P. A photoluminescence biosensor for the detection of N-acyl homoserine lactone using cysteamine functionalized ZnO nanoparticles for the early diagnosis of urinary tract infections. J. Mater. Chem. B 2020, 8, 4228–4236. [Google Scholar] [CrossRef]
  172. Abbas, S.S.; Kelly, N.L.; Patias, G.; Hanna, J.V.; McNally, T. Cysteamine functionalised reduced graphene oxide modification of maleated poly(propylene). Polymer 2020, 203, 122750. [Google Scholar] [CrossRef]
  173. Vasudevan, S.; Srinivasan, P.; Neelakantan, P.; Rayappan, J.B.B.; Solomon, A.P. Photoluminescence-Based Bioassay With Cysteamine-Capped TiO2 Nanoparticles for the Selective Recognition of N-Acyl Homoserine Lactones. Front. Bioeng. Biotechnol. 2021, 9, 750933. [Google Scholar] [CrossRef]
  174. Soares, J.A.; Ahmer, B.M.M. Detection of acyl-homoserine lactones by Escherichia and Salmonella. Curr. Opin. Microbiol. 2011, 14, 188. [Google Scholar] [CrossRef] [Green Version]
  175. Taghadosi, R.; Shakibaie, M.R.; Masoumi, S. Biochemical detection of N-Acyl homoserine lactone from biofilm-forming uropathogenic Escherichia coli isolated from urinary tract infection samples. Reports Biochem. Mol. Biol. 2015, 3, 56. [Google Scholar]
  176. Syed, M.A.; Manzoor, U.; Shah, I.; Bukhari, S.H.A. Antibacterial effects of Tungsten nanoparticles on the Escherichia coli strains isolated from catheterized urinary tract infection (UTI) cases and Staphylococcus aureus. New Microbiol. 2010, 33, 329–335. [Google Scholar] [PubMed]
  177. Matharu, R.K.; Ciric, L.; Ren, G.; Edirisinghe, M. Comparative study of the antimicrobial effects of tungsten nanoparticles and tungsten nanocomposite fibres on hospital acquired bacterial and viral pathogens. Nanomaterials. 2020, 10, 1017. [Google Scholar] [CrossRef] [PubMed]
  178. Jacobsen, S.M.; Stickler, D.J.; Mobley, H.L.T.; Shirtliff, M.E. Complicated Catheter-Associated Urinary Tract Infections Due to Escherichia coli and Proteus mirabilis. Clin. Microbiol. Rev. 2008, 21, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Uwais Bashir, M.; Ranan, D. Catheter-associated urinary tract infections. In Urinary Tract Infection: Clinical Perspectives on Urinary Tract Infection; Rané, A., Dasgupta, R., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; ISBN 1447147081. [Google Scholar]
  180. Trautner, B.W.; Darouiche, R.O. Role of biofilm in catheter-associated urinary tract infection. Am. J. Infect. Control 2004, 32, 177. [Google Scholar] [CrossRef] [PubMed]
  181. Singha, P.; Locklin, J.; Handa, H. A Review of the Recent Advances in Antimicrobial Coatings for Urinary Catheters. Acta Biomater. 2017, 50, 20. [Google Scholar] [CrossRef] [Green Version]
  182. Werneburg, G.T. Catheter-Associated Urinary Tract Infections: Current Challenges and Future Prospects. Res. Reports Urol. 2022, 14, 109. [Google Scholar] [CrossRef]
  183. Al, K.F.; Denstedt, J.D.; Daisley, B.A.; Bjazevic, J.; Welk, B.K.; Pautler, S.E.; Gloor, G.B.; Reid, G.; Razvi, H.; Burton, J.P. Ureteral Stent Microbiota Is Associated with Patient Comorbidities but Not Antibiotic Exposure. Cell Reports Med. 2020, 1, 100094. [Google Scholar] [CrossRef]
  184. Gao, L.; Wang, Y.; Li, Y.; Xu, M.; Sun, G.; Zou, T.; Wang, F.; Xu, S.; Da, J.; Wang, L. Biomimetic biodegradable Ag@Au nanoparticle-embedded ureteral stent with a constantly renewable contact-killing antimicrobial surface and antibiofilm and extraction-free properties. Acta Biomater. 2020, 114, 117–132. [Google Scholar] [CrossRef]
  185. Arkusz, K.; Paradowska, E.; Nycz, M.; Mazurek-Popczyk, J.; Baldy-Chudzik, K. Evaluation of the Antibacterial Activity of Ag- and Au-Nanoparticles Loaded TiO2 Nanotubes. J. Biomed. Nanotechnol. 2020, 16, 1416–1425. [Google Scholar] [CrossRef]
  186. Qing, Y.; Cheng, L.; Li, R.; Liu, G.; Zhang, Y.; Tang, X.; Wang, J.; Liu, H.; Qin, Y. Potential antibacterial mechanism of silver nanoparticles and the optimization of orthopedic implants by advanced modification technologies. Int. J. Nanomed. 2018, 13, 3311. [Google Scholar] [CrossRef] [Green Version]
  187. Carvalho, I.; Dias, N.; Henriques, M.; Calderon, V.S.; Ferreira, P.; Cavaleiro, A.; Carvalho, S. Antibacterial Effects of Bimetallic Clusters Incorporated in Amorphous Carbon for Stent Application. ACS Appl. Mater. Interfaces 2020, 12, 24555–24563. [Google Scholar] [CrossRef] [PubMed]
  188. Chang, C.T.; Chen, Y.T.; Hsieh, Y.K.; Girsang, S.P.; Wang, R.S.; Chang, Y.C.; Shen, S.H.; Shen, C.R.; Lin, T.P.; Wan, D.; et al. Dual-functional antibiofilm polymer composite for biodegradable medical devices. Mater. Sci. Eng. C 2021, 123, 111985. [Google Scholar] [CrossRef] [PubMed]
  189. Sajjad, S.; Uzair, B.; Shaukat, A.; Jamshed, M.; Leghari, S.A.K.; Ismail, M.; Mansoor, Q. Synergistic evaluation of AgO2 nanoparticles with ceftriaxone against CTXM and blaSHV genes positive ESBL producing clinical strains of Uro-pathogenic E. coli. IET Nanobiotechnol. 2019, 13, 433–438. [Google Scholar] [CrossRef]
  190. Farzi, S.; Ranjbar, R.; Niakan, M.; Ahmadi, M.H. Molecular Characterization of Antibiotic Resistance Associated with TEM and CTX-MESBL in Uropathogenic E. coli Strains Isolated from Outpatients. Iran. J. Pathol. 2021, 16, 386. [Google Scholar] [CrossRef] [PubMed]
  191. Gharavi, M.J.; Zarei, J.; Roshani-Asl, P.; Yazdanyar, Z.; Sharif, M.; Rashidi, N. Comprehensive study of antimicrobial susceptibility pattern and extended spectrum beta-lactamase (ESBL) prevalence in bacteria isolated from urine samples. Sci. Rep. 2021, 11, 578. [Google Scholar] [CrossRef] [PubMed]
  192. Rajivgandhi, G.; Maruthupandy, M.; Ramachandran, G.; Priyanga, M.; Manoharan, N. Detection of ESBL genes from ciprofloxacin resistant Gram negative bacteria isolated from urinary tract infections (UTIs). Front. Lab. Med. 2018, 2, 5–13. [Google Scholar] [CrossRef]
  193. Vaezi, Z.; Azizi, M.; Sadeghi Mohammadi, S.; Hashemi, N.; Naderi-Manesh, H. A novel iron quantum cluster confined in hemoglobin as fluorescent sensor for rapid detection of Escherichia coli. Talanta 2020, 218, 121137. [Google Scholar] [CrossRef]
  194. Molaabasi, F.; Sarparast, M.; Shamsipur, M.; Irannejad, L.; Moosavi-Movahedi, A.A.; Ravandi, A.; Hajipour Verdom, B.; Ghazfar, R. Shape-Controlled Synthesis of Luminescent Hemoglobin Capped Hollow Porous Platinum Nanoclusters and their Application to Catalytic Oxygen Reduction and Cancer Imaging. Sci. Rep. 2018, 8, 32918. [Google Scholar] [CrossRef] [Green Version]
  195. Sekar, R.; Basavegowda, N.; Jena, S.; Jayakodi, S.; Elumalai, P.; Chaitanyakumar, A.; Somu, P.; Baek, K.H. Recent Developments in Heteroatom/Metal-Doped Carbon Dot-Based Image-Guided Photodynamic Therapy for Cancer. Pharmaceutics 2022, 14, 1869. [Google Scholar] [CrossRef]
  196. Khalid, K.; Tan, X.; Mohd Zaid, H.F.; Tao, Y.; Lye Chew, C.; Chu, D.T.; Lam, M.K.; Ho, Y.C.; Lim, J.W.; Chin Wei, L. Advanced in developmental organic and inorganic nanomaterial: A review. Bioengineered 2020, 11, 328. [Google Scholar] [CrossRef] [Green Version]
  197. Gupta, A.; Saleh, N.M.; Das, R.; Landis, R.F.; Bigdeli, A.; Motamedchaboki, K.; Campos, A.R.; Pomeroy, K.; Mahmoudi, M.; Rotello, V.M. Synergistic antimicrobial therapy using nanoparticles and antibiotics for the treatment of multidrug-resistant bacterial infection. Nano Futur. 2017, 1, 015004. [Google Scholar] [CrossRef]
  198. Saha, B.; Bhattacharya, J.; Mukherjee, A.; Ghosh, A.K.; Santra, C.R.; Dasgupta, A.K.; Karmakar, P. In Vitro Structural and Functional Evaluation of Gold Nanoparticles Conjugated Antibiotics. Nanoscale Res. Lett. 2007, 2, 614. [Google Scholar] [CrossRef] [Green Version]
  199. Chudobova, D.; Dostalova, S.; Blazkova, I.; Michalek, P.; Ruttkay-Nedecky, B.; Sklenar, M.; Nejdl, L.; Kudr, J.; Gumulec, J.; Tmejova, K.; et al. Effect of Ampicillin, Streptomycin, Penicillin and Tetracycline on Metal Resistant and Non-Resistant Staphylococcus aureus. Int. J. Environ. Res. Public Health 2014, 11, 3233. [Google Scholar] [CrossRef] [PubMed]
  200. Bera, S.; Mondal, D. Antibacterial efficacies of nanostructured aminoglycosides. ACS Omega 2022, 7, 4724–4734. [Google Scholar] [CrossRef] [PubMed]
  201. Daghian, S.G.; Farahpour, M.R.; Jafarirad, S. Biological fabrication and electrostatic attractions of new layered silver/talc nanocomposite using Lawsonia inermis L. and its chitosan-capped inorganic/organic hybrid: Investigation on acceleration of Staphylococcus aureus and Pseudomonas aeruginosa infected wound healing. Mater. Sci. Eng. C 2021, 128, 112294. [Google Scholar] [CrossRef]
  202. Shameli, K.; Bin Ahmad, M.; Al-Mulla, E.A.J.; Shabanzadeh, P.; Bagheri, S. Antibacterial effect of silver nanoparticles on talc composites. Res. Chem. Intermed. 2015, 41, 251–263. [Google Scholar] [CrossRef]
  203. Kumar, M.S.; Ghosh, S.; Nayak, S.; Das, A.P. Recent advances in biosensor based diagnosis of urinary tract infection. Biosens. Bioelectron. 2016, 80, 497–510. [Google Scholar] [CrossRef] [PubMed]
  204. Adrover-Jaume, C.; Rojo-Molinero, E.; Clemente, A.; Russell, S.M.; Arranz, J.; Oliver, A.; De La Rica, R. Mobile origami immunosensors for the rapid detection of urinary tract infections. Analyst 2021, 145, 7916–7921. [Google Scholar] [CrossRef] [PubMed]
  205. Öztürk, R.; Murt, A. Epidemiology of urological infections: A global burden. World J. Urol. 2020, 38, 2669–2679. [Google Scholar] [CrossRef] [PubMed]
  206. Kyriakides, T.R.; Raj, A.; Tseng, T.H.; Xiao, H.; Nguyen, R.; Mohammed, F.S.; Halder, S.; Xu, M.; Wu, M.J.; Bao, S.; et al. Biocompatibility of nanomaterials and their immunological properties. Biomed. Mater. 2021, 16, 042005. [Google Scholar] [CrossRef]
  207. Najahi-Missaoui, W.; Arnold, R.D.; Cummings, B.S. Safe Nanoparticles: Are We There Yet? Int. J. Mol. Sci. 2021, 22, 385. [Google Scholar] [CrossRef] [PubMed]
  208. Anastasiadis, S.H.; Chrissopoulou, K.; Stratakis, E.; Kavatzikidou, P.; Kaklamani, G.; Ranella, A. How the Physicochemical Properties of Manufactured Nanomaterials Affect Their Performance in Dispersion and Their Applications in Biomedicine: A Review. Nanomaterials 2022, 12, 552. [Google Scholar] [CrossRef]
  209. Zhang, M.; Gao, S.; Yang, D.; Fang, Y.; Lin, X.; Jin, X.; Liu, Y.; Liu, X.; Su, K.; Shi, K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm. Sin. B 2021, 11, 2265. [Google Scholar] [CrossRef]
  210. Ahmad, F.; Salem-Bekhit, M.M.; Khan, F.; Alshehri, S.; Khan, A.; Ghoneim, M.M.; Wu, H.F.; Taha, E.I.; Elbagory, I. Unique Properties of Surface-Functionalized Nanoparticles for Bio-Application: Functionalization Mechanisms and Importance in Application. Nanomaterials 2022, 12, 1333. [Google Scholar] [CrossRef] [PubMed]
  211. Rampado, R.; Crotti, S.; Caliceti, P.; Pucciarelli, S.; Agostini, M. Recent Advances in Understanding the Protein Corona of Nanoparticles and in the Formulation of “Stealthy” Nanomaterials. Front. Bioeng. Biotechnol. 2020, 8, 166. [Google Scholar] [CrossRef] [PubMed]
  212. Gessner, A.; Lieske, A.; Paulke, B.R.; Müller, R.H. Influence of surface charge density on protein adsorption on polymeric nanoparticles: Analysis by two-dimensional electrophoresis. Eur. J. Pharm. Biopharm. 2002, 54, 165–170. [Google Scholar] [CrossRef] [PubMed]
  213. Panico, S.; Capolla, S.; Bozzer, S.; Toffoli, G.; Dal Bo, M.; Macor, P. Biological Features of Nanoparticles: Protein Corona Formation and Interaction with the Immune System. Pharmaceutics 2022, 14, 2605. [Google Scholar] [CrossRef]
  214. Nguyen, V.H.; Lee, B.J. Protein corona: A new approach for nanomedicine design. Int. J. Nanomed. 2017, 12, 3137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Gavas, S.; Quazi, S.; Karpiński, T.M. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef] [PubMed]
  216. Raafat, D.; Sahl, H.G. Chitosan and its antimicrobial potential—A critical literature survey. Microb. Biotechnol. 2009, 2, 186. [Google Scholar] [CrossRef] [Green Version]
  217. Yan, D.; Li, Y.; Liu, Y.; Li, N.; Zhang, X.; Yan, C. Antimicrobial Properties of Chitosan and Chitosan Derivatives in the Treatment of Enteric Infections. Molecules 2021, 26, 7136. [Google Scholar] [CrossRef] [PubMed]
  218. Aibani, N.; Rai, R.; Patel, P.; Cuddihy, G.; Wasan, E.K. Chitosan Nanoparticles at the Biological Interface: Implications for Drug Delivery. Pharmaceutics 2021, 13, 1686. [Google Scholar] [CrossRef] [PubMed]
  219. Akter, M.; Sikder, M.T.; Rahman, M.M.; Ullah, A.K.M.A.; Hossain, K.F.B.; Banik, S.; Hosokawa, T.; Saito, T.; Kurasaki, M. A systematic review on silver nanoparticles-induced cytotoxicity: Physicochemical properties and perspectives. J. Adv. Res. 2018, 9, 1. [Google Scholar] [CrossRef] [PubMed]
  220. Yassin, M.A.; Elkhooly, T.A.; Elsherbiny, S.M.; Reicha, F.M.; Shokeir, A.A. Facile coating of urinary catheter with bio–inspired antibacterial coating. Heliyon 2019, 5, e02986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Srisang, S.; Boongird, A.; Ungsurungsie, M.; Wanasawas, P.; Nasongkla, N. Biocompatibility and stability during storage of Foley urinary catheters coated chlorhexidine loaded nanoparticles by nanocoating: In vitro and in vivo evaluation. J. Biomed. Mater. Res. Part B Appl. Biomater. 2021, 109, 496–504. [Google Scholar] [CrossRef]
Figure 1. Mechanisms for obtaining antibiotic resistance.
Figure 1. Mechanisms for obtaining antibiotic resistance.
Nanomaterials 13 00555 g001
Figure 2. Antimicrobial therapy strategies.
Figure 2. Antimicrobial therapy strategies.
Nanomaterials 13 00555 g002
Figure 3. Diagnosis of urinary tract infections.
Figure 3. Diagnosis of urinary tract infections.
Nanomaterials 13 00555 g003
Figure 4. Biofilm infections in the urinary tract.
Figure 4. Biofilm infections in the urinary tract.
Nanomaterials 13 00555 g004
Figure 5. Nanoparticles for treating biofilm infections in the urinary tract.
Figure 5. Nanoparticles for treating biofilm infections in the urinary tract.
Nanomaterials 13 00555 g005
Figure 6. Nanoparticles—common mechanisms of action against UTIs.
Figure 6. Nanoparticles—common mechanisms of action against UTIs.
Nanomaterials 13 00555 g006
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

Crintea, A.; Carpa, R.; Mitre, A.-O.; Petho, R.I.; Chelaru, V.-F.; Nădășan, S.-M.; Neamti, L.; Dutu, A.G. Nanotechnology Involved in Treating Urinary Tract Infections: An Overview. Nanomaterials 2023, 13, 555. https://doi.org/10.3390/nano13030555

AMA Style

Crintea A, Carpa R, Mitre A-O, Petho RI, Chelaru V-F, Nădășan S-M, Neamti L, Dutu AG. Nanotechnology Involved in Treating Urinary Tract Infections: An Overview. Nanomaterials. 2023; 13(3):555. https://doi.org/10.3390/nano13030555

Chicago/Turabian Style

Crintea, Andreea, Rahela Carpa, Andrei-Otto Mitre, Robert Istvan Petho, Vlad-Florin Chelaru, Sebastian-Mihail Nădășan, Lidia Neamti, and Alina Gabriela Dutu. 2023. "Nanotechnology Involved in Treating Urinary Tract Infections: An Overview" Nanomaterials 13, no. 3: 555. https://doi.org/10.3390/nano13030555

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