Next Article in Journal
The Safe Values of Quantitative Perfusion Parameters of ICG Angiography Based on Tissue Oxygenation of Hyperspectral Imaging for Laparoscopic Colorectal Surgery: A Prospective Observational Study
Next Article in Special Issue
Synergy Assessment of Four Antimicrobial Bioactive Compounds for the Combinational Treatment of Bacterial Pathogens
Previous Article in Journal
Functional State of the Motor Centers of the Lumbar Spine after Contusion (Th8-Th9) with Application of Methylprednisolone-Copolymer at the Site of Injury
Previous Article in Special Issue
A CAF-Based Two-Cell Hybrid Co-Culture Model to Test Drug Resistance in Endometrial Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Computational Guided Drug Targets Identification against Extended-Spectrum Beta-Lactamase-Producing Multi-Drug Resistant Uropathogenic Escherichia coli

Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(7), 2028; https://doi.org/10.3390/biomedicines11072028
Submission received: 28 June 2023 / Revised: 14 July 2023 / Accepted: 15 July 2023 / Published: 19 July 2023

Abstract

:
Urinary tract infections (UTIs) are one of the most frequent bacterial infections in the world, both in the hospital and community settings. Uropathogenic Escherichia coli (UPEC) are the predominant etiological agents causing UTIs. Extended-spectrum beta-lactamase (ESBL) production is a prominent mechanism of resistance that hinders the antimicrobial treatment of UTIs caused by UPEC and poses a substantial danger to the arsenal of antibiotics now in use. As bacteria have several methods to counteract the effects of antibiotics, identifying new potential drug targets may help in the design of new antimicrobial agents, and in the control of the rising trend of antimicrobial resistance (AMR). The public availability of the entire genome sequences of humans and many disease-causing organisms has accelerated the hunt for viable therapeutic targets. Using a unique, hierarchical, in silico technique using computational tools, we discovered and described potential therapeutic drug targets against the ESBL-producing UPEC strain NA114. Three different sets of proteins (chokepoint, virulence, and resistance genes) were explored in phase 1. In phase 2, proteins shortlisted from phase 1 were analyzed for their essentiality, non-homology to the human genome, and gut flora. In phase 3, the further shortlisted putative drug targets were qualitatively characterized, including their subcellular location, broad-spectrum potential, and druggability evaluations. We found seven distinct targets for the pathogen that showed no similarity to the human proteome. Thus, possibilities for cross-reactivity between a target-specific antibacterial and human proteins were minimized. The subcellular locations of two targets, ECNA114_0085 and ECNA114_1060, were predicted as cytoplasmic and periplasmic, respectively. These proteins play an important role in bacterial peptidoglycan biosynthesis and inositol phosphate metabolism, and can be used in the design of drugs against these bacteria. Inhibition of these proteins will be helpful to combat infections caused by MDR UPEC.

Graphical Abstract

1. Introduction

The World Health Organization (WHO) has listed AMR as one of the top ten worldwide public health concerns to humanity in the twenty-first century [1]. Recently, AMR has been referred to as the “silent tsunami facing modern medicine” [2]. The heightened use/misuse of antibiotics in human medicine and animal agriculture primarily contributes to this phenomenon. An alarming increase of AMR in bacteria causes community or hospital-acquired infections. Of particular interest is the multi-drug resistant (MDR) pathogen Escherichia coli (E. coli), which is the most common causative pathogen in many types of infections, especially in countries with poor healthcare systems [3]. E. coli are part of normal human and animal gut flora, but are also the predominant cause of community and hospital-acquired UTIs. AMR among UPEC has significantly grown recently, restricting the choice of treatment options. Special notes are ESBL-producing E. coli (ESBL-EC), which are responsible for severe human morbidity and mortality, with significant economic losses and disease burdens [4]. ESBLs are a fast-growing class of beta-lactamases that can hydrolyze oxy-imino cephalosporins (ceftriaxone, cefuroxime, ceftazidime, cefepime, and cefotaxime) and monobactams which results in drug resistance [5]. Thus, these bacteria have become resistant to many available antibiotics, and may also be resistant to drugs of last resort such as carbapenems [6]. The use of carbapenems has dramatically increased over the previous few years, leading to carbapenem-resistant Enterobacteriaceae (CRE), against which hardly any antibiotics are available, except for colistin [6]. ESBLs are rampant in low and middle-income countries (LMICs) [4]. WHO has put ESBL-EC on its priority list against which new therapeutics are being developed [7].
The accessibility of both the human and microbial genomes has made it easier to apply comparative and subtractive genomics approaches, in which the genomes of the human host and pathogens are compared and homologous host proteins are identified as non-targets. Pathogen-specific proteins with various therapeutic properties have been discovered [8,9,10]. Such approaches have been used to identify non-homologous proteins as potential drug targets in many pathogens, including Staphylococcus saprophyticus, Mycobacterium avium, Acinetobacter baumannii, Pseudomonas aeruginosa, Burkholderia pseudomallei, and Streptococcus pneumoniae [9,11,12,13]. These approaches, combined with the advancements in computational biology and the availability of diverse bioinformatics tools, are revolutionizing fields of drug discovery and design by minimising time and expense of wet-lab screening [13,14].
In this study, we applied a subtractive genomics approach using an ESBL- producing MDR UPEC strain NA114. This UPEC strain was identified in Pune, India, in the urine of a 70-year-old man who had prostatitis. It is an MDR pathogen and ESBL-producing strain, resistant to amoxicillin, co-trimoxazole, tetracycline, gentamicin, nalidixic acid, ciprofloxacin, ceftazidime, and nitrofurantoin [15]. To identify and select the proteins that are crucial for the pathogen’s survival, we made use of a variety of computational techniques, software, and web databases. Only proteins specific to pathogens were shortlisted, thereby avoiding host homology. In order to identify possible broad-spectrum therapeutic targets, we considered all functional metabolic pathways, virulence genes, and resistance genes and discovered seven distinct targets that are unique to UPEC. Furthermore, as they showed no similarity to the human proteome, the possibilities of cross-reactivity between a new drug that uses one of these targets and human proteins were minimized. The subcellular locations of two targets, ECNA114_0085 and ECNA114_1060, were predicted as cytoplasmic and periplasmic, respectively. These proteins play an important role in peptidoglycan biosynthesis and inositol phosphate metabolism. Therefore, the development of new drugs against these targets could be promising steps towards eliminating UTIs caused by E. coli.

2. Material and Methods

Figure 1 displays the full workflow that was employed in this study.

2.1. Phase I: Comparative Analysis of Pathogen and Human Proteome

Step1: Non-Homology Analysis

The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used to compare the pathways of the host Homo sapiens (H. sapiens) and the UPEC strain NA114 [16]. Pathways that are not present in the human host were considered to be distinct pathways. All proteins in the pathways were searched using the protein basic local alignment search tool (BLASTp) against the non-redundant database. Non-homologous proteins were chosen based on rigorous measurements showing no similarity [17].

2.2. Chokepoint Analysis

In a metabolic network, a “chokepoint reaction” is a reaction that consumes or creates a single substrate or product. The inhibition of enzymes involved in chokepoint reaction will hamper the important cell function [18]. Chokepoint analysis is carried out using simple statistics. The confidence level (CL) of all non-homologous proteins (enzymes) has been calculated by using the simple formula [19],
C o n f i d e n c e   l e v e l   o f   a n   e n z y m e = N o .   o f   p a t h w a y s   i n   w h i c h   a n   e n z y m e   i s   f o u n d   a s   a   c h o k e   p o i n t / T o t a l   n o .   o f   p a t h w a y s   o f   t h e   e n z y m e × 100 .
Proteins with confidence scores below 50% were removed, while those with confidence scores above 50% were chosen for further study.

2.2.1. Step 2: Analysis of Virulence Genes

Virulence factors (VF) have been identified as prospective therapeutic targets in drug development. The pathogen would become avirulent if these virulence proteins were inhibited, as these proteins are essential for the establishment and severity of infection [20]. The virulence factor database (VFDB) contains virulence genes from different bacterial species, including E. coli [21]. The VFDB was used to compile a comprehensive list of virulence genes, and corresponding sequences of these genes were taken from the national center for biotechnology information (NCBI)/KEGG database.

2.2.2. Step 3: Analysis of Resistance Genes

In a survey in India, resistance rates of UPEC to various antibiotics were reported for gentamicin (58.2%), beta-lactams (57.4%), quinolones (74.5%), amikacin (33.4%), nalidixic acid (77.7%), co-trimoxazole (48.5%) and cefuroxime (56%) [22]. The genes that produce resistance to popular antibiotics were found using the antibiotic resistance database (ARDB). High-confidence interactors were predicted using the Search Technique for the Retrieval of Interacting Genes/Proteins (STRING) version 10 for each resistant protein [23]. Further, the proteins with high interactions were manually screened for their presence in the human host.

2.3. Phase II: Subtractive Analysis

2.3.1. Analysis of Essential Genes

We identified potential therapeutic drug targets for powerful essential proteins using a BLASTp search against the database of essential genes (DEG) [24]. DEG, a database of essential genes (genes required for an organism’s survival), contains essential genes from both eukaryotic and prokaryotic organisms. Protein alignments with an e-value of 0.005 or less were considered more meaningful. The resultant proteins were reconstituted and used as input for the non-homology analysis.

2.3.2. Non-homology Analysis

The goal of the non-homology study is to identify proteins specific to the pathogen that are non-homologous to the human host. This process is important because it prevents the drug from binding to any homologous host proteins and reduces the drug’s undesired cross-reactivity [25]. All short-listed proteins were analysed through a BLASTp against a non-redundant H. sapiens database [17,25,26]. Proteins not present in the human host were chosen for the following phase.

2.3.3. Human Gut Flora Non-homology Analysis

Shortlisted proteins from the prior step were compared to the proteome of human gut microflora. A normal healthy human’s gastrointestinal tract contains approximately 1014 bacterial species [27]. The gut microbiota has a symbiotic relationship with the host, and it plays an important role in metabolism by digesting food particles and protecting the gut against harmful bacteria invasion [28]. Unintentional inhibition of gut flora proteins can degrade the microbiota, resulting in negative consequences for the host. To avoid pharmacological cross-reactivity with gut microflora proteins, the target proteins were homologized against the gut flora proteome using BLASTp with an expected value (e-value) of 0.005 [26]. The analysis included a list of gut microorganisms reported in the literature [29]. The proteins with no more than ten matches on their own were chosen. The potential target proteins were further analyzed for their quantitative characterization.

2.4. Phase III: Quantitative Characterization of Putative Drug Targets

2.4.1. Subcellular Localization Prediction

E.coli is a gram-negative bacterium that possesses an outer cell membrane. Therefore, proteins after synthesis were localized in five possible locations, namely, cytoplasm, periplasm, and the plasma, extracellular, and outer membranes. The localization analysis aims to identify a protein as a potential therapeutic or vaccination target. Surface membrane proteins can be employed as vaccination targets, while cytoplasmic/periplasmic proteins can be exploited as therapeutic drug targets [30]. Once the potential targets were selected, identification of the location of these proteins was attempted to know their functional assignment. For the identification of sub-cellular locations, the Cell Ontology-based Classification (Cello) tool [31], PSORTb (https://www.psort.org/psortb/) [32], and a program for identification of sub-cellular localization of bacterial proteins (ProtCompB) were used.

2.4.2. Broad-Spectrum Analysis

For the identification of broad-spectrum targets, potential therapeutic targets were examined using a BLASTp search against a large number of pathogenic microorganisms [29]. A list of pathogenic bacteria involved in UTIs reported in the virulence factor database (VFDB), and pathogenic bacteria from the literature, as reported by Shanmugham et al. 2013 were considered in this analysis [21]. According to the homology analysis for each pathogen, close homologs are more likely to represent a “promising broad-spectrum target” if they are found in a larger number of pathogenic species.

2.4.3. Druggability Analysis

Further, we performed a BLASTp search against the DrugBank database with an e-value of 0.005 to assess each protein’s drug-ability potential. The Drug Bank database is a one-of-a-kind resource that combines pharmacological data with information on drug targets at the sequencing, structure, and pathway levels [33].

3. Results

3.1. Phase I: Comparative Analysis

The metabolic pathways of the host H. sapiens and the UPEC have been compared in silico. Pathways that are not found in humans but are found in pathogens are referred to as distinct pathways. The 119 metabolic pathways considered in this study are linked to nucleotide, carbohydrate, amino acid, or vitamin metabolism, or cofactor biosynthesis. Thirty four of the 119 pathways have been identified as distinct. Non-homologous proteins were identified in 30 of the 34 pathways.

3.1.1. Chokepoint Enzymes

Chokepoint analysis was investigated using non-homologous proteins from several metabolic pathways. Thirty-one of the 62 proteins were identified as choke point enzymes, with a confidence level of more than 50%, and were chosen for further analysis (Table 1).

3.1.2. Virulence Factors Analysis

Several types of virulence factors, including adherence, iron uptake-aerobactin, and hemolysin are enlisted in VFDB. In UPEC, 69 virulence genes were found. Out of 69, nineteen were found in UPEC strain NA114 (Table 2).

3.1.3. Resistance Gene Analysis

Resistance genes were taken from the ARDB. In the UPEC strain used in this study, we found 1805 resistance genes. These proteins were further screened from the STRING database, which resulted in 20 genes with high interactions. Out of these 20, six proteins were present in UPEC and not found in humans (Table 3). Protein sequences of these resistance-causing proteins were taken from KEGG/NCBI database. Generally, resistance-causing proteins and their related proteins are thought to be ‘promising therapeutic targets’, because inhibiting them could stop the drug resistance mechanism from working.

3.2. Phase II: Subtractive Channel of Analysis

3.2.1. Analysis of Essential Genes

Using the DEG server, and a threshold e-value of 0.005, the combined proteins from phase 1 were further screened for essential genes. The most important condition for a prospective therapeutic target is that it must be a protein that the organism needs to survive. Fifteen of the 52 input proteins identified in phase I were found to be required for the pathogen’s survival and growth (Table 4). Proteins that did not match against any in the DEG database were regarded as non-essential and excluded from the study. The selectivity/specificity of the proteins were analyzed by finding proteins that are non-homologous to the human and gut microbiota proteome.

3.2.2. Non-homology Analysis

The use of proteins that are homologous to the host as therapeutic targets could hurt the host’s metabolism. As a result, several in silico drug-target-discovery algorithms involving filtering out proteins homologous to the human proteome were utilized as the initial step. Non-homology analysis was performed on the short-listed protein datasets. BLASTp was used to look for similarities between the fifteen UPEC proteins and the entire proteome of H. sapiens (host). Five proteins were found to be homologous to a human host and excluded from the study. The remaining ten non-homologous proteins were used in further studies.

3.2.3. Gut Flora Non-homology Analysis

The resulting list of proteins that are non-homologous to H. sapiens was screened against the whole proteome microbiota and other gut flora organisms found in the literature using the BLASTp search [29]. From gut flora non-homology analysis, three proteins were found as homologs in microbiota and other gut flora organisms and excluded from the study. The remaining seven proteins were further assessed as putative drug targets.

3.3. Phase III

In this phase, the putative drug targets were further analyzed for their properties, namely, cellular location, broad-spectrum, and druggability analysis.

3.3.1. Subcellular Localization Prediction of Putative Targets

The sub-cellular location was predicted by using ProtCompB, Cello, and PSORTb tools. ProtCompB predicts the localization of proteins at the subcellular level in gram-negative bacteria; by using these, five of the seven proteins were found in the inner membrane, one in the periplasm, and one in the cytoplasm. Cello predicts the localization of protein domains using a multi-class support vector machine (SVM) system based on the physicochemical properties of proteins and predicted the location of one protein in the cytoplasm, one in the periplasm, and five in the inner membrane. PSORTb is another tool for the prediction of protein location for gram-positive strains, gram-negative strains, and archaeal sequences. In our dataset, it returned five proteins as cytoplasmic-membrane, and two as cytoplasm. By comparing the results from different tools, the location of five proteins was found in the inner membrane, one in the cytoplasm and one in the periplasm (Table 5).

3.3.2. Broad-Spectrum Analysis

Comparative genomic analysis of the screened targets, using a clinically significant E. coli strain that causes UTIs described in the VFDB as reference, allowed for an efficient assessment of potential broad-spectrum therapy targets. The broad-spectrum investigation used a list of 240 pathogenic pathogens. A promising broad-spectrum target was discovered using a BLASTp homology search against the entire proteome of each of these bacterial pathogens. The homology search revealed that the screened targets had close homologs in multiple pathogenic species, indicating that E. coli proteins are multispecies. Therapeutic compounds that inhibit such broad-spectrum targets could help to eradicate UPEC infections and could be used as possible UPEC-specific drug targets. Such pathogen-specific targets could help to lower the possibility of treatment resistance in a variety of infections. All proteins had more than 70 matches, indicating that the therapeutic targets have a broad spectrum.

3.3.3. Druggability Analysis

The druggability of the short-listed candidate drug targets was assessed by utilizing a sequence-similarity search against the Drug Bank target database. All seven potential drug targets were searched against the Drug Bank target database manually, which includes US Food and Drug Administration (FDA)-approved drugs, revealing that ECNA114_0085 is homologous to a known target, D-Alanine-D-Alanine-ligase. None of the other targets exhibited any matches to the drug target database (Table 6).

4. Discussion

In this study, we used a broad approach and included all functional biosynthetic pathways, such as the metabolism of carbohydrates, energy, amino acids, vitamins, terpenoids, and ketides, as well as those for the biodegradation of xenobiotics. We identified distinct non-homologous pathways that are only found in pathogens and not in human hosts, and the proteins found in these pathways were classified as distinct proteins [34]. Out of 34 considered pathways, non-homologous proteins were found in 30 pathways. However, the fact that they are distinct from, or unrelated to human hosts does not qualify them as effective therapeutic drug targets, as isoenzymes or paralogs exist [35]. To remove this bias, a chokepoint analysis was done. A chokepoint reaction consumes or creates a particular product [18]. Therefore, inhibiting an enzyme that uses a harmful substrate may result in the accumulation of toxic products. Alternatively, if the inhibited enzyme is involved in the creation of a special product, inhibiting it would starve the cell, which may ultimately impair vital cell processes [36]. In light of this, enzymes implicated in chokepoint reactions may be crucial for infectivity and thus, may represent a potential therapeutic target for drug discovery. Thirty-one enzymes from the 62 non-homologous enzymes identified in various metabolic pathways were recognized as chokepoint enzymes. In this work, we have also taken UPEC virulence factors into consideration. Targeting virulence factors has two benefits: first, it will reduce pathogenicity; and second, selection pressure will be minimized, preventing the development of drug resistance. Cinnamaldehyde, baicalein, naringenin, and catechin are a few of the recently identified anti-virulence substances that act by preventing the initiation of quorum sensing (QS) and biofilm production [37,38]. In our study, UPEC strain NA114 was found to contain 19 virulence factors, including those involved in lysine degradation, ABC transporters, biosynthesis of siderophores, two-component pathways, and bacterial secretion systems. By stymieing the actions of proteins linked to antibiotic resistance proteins, drug resistance can also be avoided. To identify potential therapeutic targets for UPEC NA114, we thus included antibiotic resistance genes as targets in the current investigation. Similar combinatorial approaches have been used for the identification of putative drug targets for Mycobacterium abscessus where 40 targets were identified [29]. Out of 15 proteins found in the DEG database deemed to be essential for the survival of UPEC, further non-homology and gut flora non-homology analysis yielded seven putative drug targets (Table 3). Since the eligibility and effectiveness of a protein as a therapeutic target depends on where it is located in the cell, in silico tools were employed to predict the subcellular location of the protein [13,39]. Proteins found in the membrane are not ideal as therapeutic targets due to their difficulty in purification and testing. We found two potent druggable proteins (ECNA114_0085 and ECNA114_1060) as promising targets in UPEC strain NA114.
The cytoplasmic ECNA114_0085 protein, identified as a potential target here, is a D-alanine-D-alanine ligase (Ddl) involved in the synthesis of peptidoglycan [9,40]. The peptidoglycan precursor UDPMurNAc-pentapetide contains the terminal dipeptide D-Ala-D-Ala, which is an essential building component for peptidoglycan cross-linking, which provides stability to the cell wall. Therefore, blocking the enzyme Ddl can dramatically reduce the strength of the cell wall, which causes the bacteria to be osmotically lysed. According to a prior study, Ddl is an attractive target for the discovery of drugs in Mycobacterium tuberculosis [41]. Vancomycin, which acts only on gram-positive bacteria, binds to Ddl, which inhibits glucosyltransferase, thereby preventing the polymerization and synthesis of N-acetylglucosamine (NAG) and N-acetylmuramic acid (NAM) throughout the peptidoglycan layer [42]. The essential penicillin-binding proteins (PBPs), which are enzymes involved in the final stages of peptidoglycan cross-linking in both gram-negative as well as gram-positive bacteria, covalently bind to beta-lactam antibiotics, making them bactericidal substances that prevent the formation of bacterial cell walls. Early intracellular phases of cell wall synthesis have received little attention as prospective therapeutic targets so far, in contrast to the extracellular stages of peptidoglycan biosynthesis, which are inhibited by beta-lactam and glycopeptide antibiotics [43]. Currently, only two drugs are in use to block intracellular peptidoglycan synthesis: D-cycloserine, which blocks both Ddl and alanine racemase, and fosfomycin, a MurA ligase inhibitor. The internal peptidoglycan precursor UDPMurNAc-pentapeptide is assembled by the mur ligases MurC, MurD, MurE, and MurF by the sequential addition of L-Ala, D-Glu, m-Dpm or L-Lys, and D-Ala-D-Ala to UDP-MurNAc. Ddl is responsible for supplying the MurF substrate, D-alanyl-D-alanine [43]. Because cross-linking of peptidoglycan chains takes place between the penultimate D-Ala in a second pentapeptide strand and the C6-NH2-group of meso-diaminopimelic acid in gram-negative bacteria and the NH2-group of pentaglycine in gram-positive Staphylococcus, this terminal dipeptide is crucial in the construction of the bacterial cell wall. In both situations, the intrastrand D-Ala-D-Ala bond is broken and a new interstrand peptide bond is formed [43]. This enzyme is common in prokaryotes, but absent in eukaryotes, making it a likely candidate target for antibiotic development. It is possible to design inhibitors against this enzyme, crucial to the peptidoglycan synthesis process, that could cause a loss in the structural integrity of bacterial cell walls and osmotic lysis of pathogenic bacteria.
The periplasmic protein appA (ECNA114_1060) involved in the metabolism of inositol phosphate was found to be another attractive target. Gram-negative bacteria are extremely resistant to the penetration of antimicrobials due to their double-layered cell envelope and a variety of efflux pumps. Proteins in the periplasm are easier to target, since only penetration of the outer membrane is required, whereas cytoplasmic targets are difficult to reach [44]. In addition to assisting in the production of virulence factors, adhesion molecules, and signaling molecules, periplasmic proteins are crucial for sustaining cell viability, cell division, and membrane integrity. All eukaryote genomes contain the inositol polyphosphate production enzymes, yet standard search techniques frequently fail to uncover the amino acid sequence homology of these enzymes [45]. Similarity searching between microbial and human inositol phosphate kinases is restricted to fewer catalytically important residues, which include proline, aspartic acid, lysine, glycine, serine, leucine, tyrosine, isoleucine, histidine, threonine, glutamic acid, and phenylalanine [45]. Recent studies of the Inositol phosphate metabolic pathways in pathogenic protozoa (Trypanosome brucei) and fungi (Cryptococcus neoformans) are being exploited as promising antimicrobial targets [45]. Inositol phosphate metabolism has been identified as a putative drug target for many bacterial pathogens including Staphylococcus saprophyticus, Klebsiella pneumoniae, and Streptococcus pneumoniae [11,46,47]. Despite the benefits and optimistic future of comparative genomics in the identification of possible therapeutic targets, there are still certain limitations. Despite the fact that comparative genomics is frequently utilized in the creation of medications to combat drug-resistant bacteria, the failure rate of existing antibiotics is substantially higher than the rate at which new antibiotics are developed. Additionally, antibiotics are transient treatments for infections. The use of comparative genomics in the creation of antibiotics is a long-debated topic due to the fact that their value is significantly lower than medications used in the treatment of chronic diseases [48]. A further problem is that, despite the fact that comparative genomics can decrease the number of experimental targets and identify some appealing proteins as prospective therapeutic targets, the range of potential targets that can be screened by this method is still quite broad and the cost and time used in the development process is prohibitive. The majority of these putative targets identified by comparative genomics are unlikely to be validated through experiments. Therefore, it would be advantageous to integrate network-based approaches with comparative genomics to further limit the range of experimental targets. This will use less time and material, and cut costs during the early stages of drug research and development.

5. Conclusions

Using a unique hierarchical in silico technique, we discovered and described two potent druggable proteins as prospective therapeutic drug targets against ESBL-producing MDR UPEC strain NA114. The identified drug targets are distinct and have the potential to be exploited for designing new antimicrobial agents against UPEC. The computer-aided drug design method can also be utilized to identify homologous compounds for these targets. The search for possible therapeutic targets in ESBL-producing MDR UPEC strain NA114 was directed by comparative and subtractive genomics. Essentiality, non- homology to the human host, availability in the drug bank, and sub- cellular location were used to narrow down the list of targets in this study. Our research uncovered prospective therapeutic targets (ECNA114_0085 and ECNA114_1060) that are critical in the treatment of E. coli urinary tract infections.

Author Contributions

H.K. and N.T., Conceptualization. H.K., Data curation. H.K., Formal analysis. H.K., Investigation. H.K., V.M. and N.C., Methodology. H.K., Software. B.M. and N.T., Supervision. H.K. and N.T., Validation. H.K. and N.T. Writing—original draft. H.K. and N.T., Writing—review & editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We gratefully acknowledge to the Director, PGIMER Chandigarh for providing facilities to carry out this work. The Indian Council of Medical Research (ICMR), New Delhi support in the form of a fellowship (ICMR/RA/Fellowship/2021/11125) to the first author is highly acknowledged.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murray, C.J.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Robles Aguilar, G.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  2. Exner, M.; Bhattacharya, S.; Christiansen, B.; Gebel, J.; Goroncy-Bermes, P.; Hartemann, P.; Heeg, P.; Ilschner, C.; Kramer, A.; Larson, E.; et al. Antibiotic Resistance: What Is so Special about Multidrug-Resistant Gram-Negative Bacteria ? Antibiotikaresistenz: Was Ist so Besonders an Den Gram-Negativen. GMS Hyg. Infect. Control 2017, 12, Doc05. [Google Scholar]
  3. Kayastha, K.; Dhungel, B.; Karki, S.; Adhikari, B.; Banjara, M.R.; Rijal, K.R.; Ghimire, P. Extended-Spectrum β-Lactamase-Producing Escherichia Coli and Klebsiella Species in Pediatric Patients Visiting International Friendship Children’s Hospital, Kathmandu, Nepal. Infect. Dis. Res. Treat. 2020, 13, 117863372090979. [Google Scholar] [CrossRef] [Green Version]
  4. Taneja, N.; Rao, P.; Arora, J.; Dogra, A. Occurrence of ESBL & Amp-C b -Lactamases & Susceptibility to Newer Antimicrobial Agents in Complicated UTI. Indian J Med Res. 2008, 127, 85–88. [Google Scholar]
  5. Shaikh, S.; Fatima, J.; Shakil, S.; Rizvi, S.M.D.; Kamal, M.A. Antibiotic Resistance and Extended Spectrum Beta-Lactamases: Types, Epidemiology and Treatment. Saudi J. Biol. Sci. 2015, 22, 90–101. [Google Scholar] [CrossRef] [PubMed]
  6. Kaza, P.; Mahindroo, J.; Veeraraghavan, B.; Mavuduru, R.S.; Mohan, B.; Taneja, N. Evaluation of Risk Factors for Colistin Resistance among Uropathogenic Isolates of Escherichia Coli and Klebsiella Pneumoniae: A Case–Control Study. J. Med. Microbiol. 2019, 68, 837–847. [Google Scholar] [CrossRef] [PubMed]
  7. Asokan, G.V.; Ramadhan, T.; Ahmed, E.; Sanad, H. WHO Global Priority Pathogens List: A Bibliometric Analysis of Medline-Pubmed for Knowledge Mobilization to Infection Prevention and Control Practices in Bahrain. Oman. Med. J. 2019, 34, 184–193. [Google Scholar] [CrossRef] [PubMed]
  8. Chakrabarty, R.P.; Alam, A.S.M.R.U.; Shill, D.K.; Rahman, A. Identification and Qualitative Characterization of New Therapeutic Targets in Stenotrophomonas Maltophilia through in Silico Proteome Exploration. Microb. Pathog. 2020, 149, 104293. [Google Scholar] [CrossRef] [PubMed]
  9. Kaur, H.; Kalia, M.; Taneja, N. Identification of Novel Non-Homologous Drug Targets against Acinetobacter Baumannii Using Subtractive Genomics and Comparative Metabolic Pathway Analysis. Microb. Pathog. 2021, 152, 104608. [Google Scholar] [CrossRef]
  10. Kaur, H.; Kalia, M.; Singh, V.; Modgil, V.; Mohan, B.; Taneja, N. In Silico Identification and Characterization of Promising Drug Targets in Highly Virulent Uropathogenic Escherichia Coli Strain CFT073 by Protein-Protein Interaction Network Analysis. Inf. Med. Unlocked 2021, 25, 100704. [Google Scholar] [CrossRef]
  11. Shahid, F.; Ashfaq, U.A.; Saeed, S.; Munir, S.; Almatroudi, A.; Khurshid, M. In Silico Subtractive Proteomics Approach for Identification of Potential Drug Targets in Staphylococcus Saprophyticus. Int. J. Environ. Res. Public Health 2020, 17, 3644. [Google Scholar] [CrossRef] [PubMed]
  12. Solanki, V.; Tiwari, V. Subtractive Proteomics to Identify Novel Drug Targets and Reverse Vaccinology for the Development of Chimeric Vaccine against Acinetobacter Baumannii. Sci. Rep. 2018, 8, 9044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Dutta, A.; Singh, S.K.; Ghosh, P.; Mukherjee, R.; Mitter, S.; Bandyopadhyay, D. In Silico Identification of Potential Therapeutic Targets in the Human Pathogen Helicobacter Pylori. Silico Biol. 2006, 6, 43–47. [Google Scholar]
  14. Lin, X.; Li, X.; Lin, X. A Review on Applications of Computational Methods in Drug Screening and Design. Molecules 2020, 25, 1375. [Google Scholar] [CrossRef] [Green Version]
  15. Avasthi, T.S.; Kumar, N.; Baddam, R.; Hussain, A.; Nandanwar, N.; Jadhav, S.; Ahmed, N. Genome of Multidrug-Resistant Uropathogenic Escherichia Coli Strain NA114 from India. J. Bacteriol. 2011, 193, 4272–4273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kanehisa, M.; Goto, S.; Kawashima, S.; Nakaya, A. The KEGG Databases at GenomeNet. Nucleic Acids Res. 2002, 30, 42–46. [Google Scholar] [CrossRef] [Green Version]
  17. Anishetty, S.; Pulimi, M.; Pennathur, G. Potential Drug Targets in Mycobacterium Tuberculosis through Metabolic Pathway Analysis. Comput. Biol. Chem. 2005, 29, 368–378. [Google Scholar] [CrossRef]
  18. Singh, S.; Malik, B.K.; Sharma, D.K. Choke Point Analysis of Metabolic Pathways in E.Histolytica: A Computational Approach for Drug Target Identification. Bioinformation 2007, 2, 68–72. [Google Scholar] [CrossRef]
  19. Sc, S.K.M.; Phil, M. Protocol of Rice Genome Annotation through Comparative Functional Genomics Approach. Genome 2009, 4, 1–8. [Google Scholar]
  20. Baron, C.; Coombes, B. Targeting Bacterial Secretion Systems: Benefits of Disarmament in the Microcosm. Infect. Disord. Drug Targets 2007, 7, 19–27. [Google Scholar] [CrossRef]
  21. Chen, L.; Zheng, D.; Liu, B.; Yang, J.; Jin, Q. VFDB 2016: Hierarchical and Refined Dataset for Big Data Analysis--10 Years On. Nucleic Acids Res. 2016, 44, D694–D697. [Google Scholar] [CrossRef] [PubMed]
  22. Niranjan, V.; Malini, A. Antimicrobial Resistance Pattern in Escherichia Coli Causing Urinary Tract Infection among Inpatients. Indian J. Med. Res. 2014, 139, 945–948. [Google Scholar] [PubMed]
  23. Szklarczyk, D.; Franceschini, A.; Wyder, S.; Forslund, K.; Heller, D.; Huerta-Cepas, J.; Simonovic, M.; Roth, A.; Santos, A.; Tsafou, K.P.; et al. STRING V10: Protein–Protein Interaction Networks, Integrated over the Tree of Life. Nucleic Acids Res. 2015, 43, D447–D452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Luo, H.; Lin, Y.; Gao, F.; Zhang, C.T.; Zhang, R. DEG 10, an Update of the Database of Essential Genes That Includes Both Protein-Coding Genes and Noncoding Genomic Elements. Nucleic Acids Res 2014, 42, D574–D580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Sarkar, M.; Maganti, L.; Ghoshal, N.; Dutta, C. In Silico Quest for Putative Drug Targets in Helicobacter Pylori HPAG1: Molecular Modeling of Candidate Enzymes from Lipopolysaccharide Biosynthesis Pathway. J. Mol. Model 2012, 18, 1855–1865. [Google Scholar] [CrossRef]
  26. Altschul, S.F.; Gish, W.; Miller, W.; Myers, E.W.; Lipman, D.J. Basic Local Alignment Search Tool. J. Mol. Biol. 1990, 215, 403–410. [Google Scholar] [CrossRef]
  27. Fujimura, K.E.; Slusher, N.A.; Cabana, M.D.; Lynch, S.V. Role of the Gut Microbiota in Defining Human Health. Expert Rev. Anti. Infect. Ther. 2010, 8, 435–454. [Google Scholar] [CrossRef] [Green Version]
  28. Rabizadeh, S.; Sears, C. New Horizons for the Infectious Diseases Specialist: How Gut Microflora Promote Health and Disease. Curr. Infect. Dis. Rep. 2008, 10, 92–98. [Google Scholar] [CrossRef] [Green Version]
  29. Shanmugham, B.; Pan, A. Identification and Characterization of Potential Therapeutic Candidates in Emerging Human Pathogen Mycobacterium Abscessus: A Novel Hierarchical In Silico Approach. PLoS ONE 2013, 8, e59126. [Google Scholar] [CrossRef]
  30. Barh, D.; Tiwari, S.; Jain, N.; Ali, A.; Santos, A.R.; Misra, A.N.; Azevedo, V.; Kumar, A. In Silico Subtractive Genomics for Target Identification in Human Bacterial Pathogens. Drug Dev. Res. 2011, 72, 162–177. [Google Scholar] [CrossRef]
  31. Yu, C.; Lin, C.; Hwang, J.-K. Predicting Subcellular Localization of Proteins for Gram-Negative Bacteria by Support Vector Machines Based on n -Peptide Compositions. Proteins Sci. 2004, 13, 1402–1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Yu, N.Y.; Wagner, J.R.; Laird, M.R.; Melli, G.; Rey, S.; Lo, R.; Dao, P.; Cenk Sahinalp, S.; Ester, M.; Foster, L.J.; et al. PSORTb 3.0: Improved Protein Subcellular Localization Prediction with Refined Localization Subcategories and Predictive Capabilities for All Prokaryotes. Bioinformatics 2010, 26, 1608–1615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wishart, D.S.; Knox, C.; Guo, A.C.; Cheng, D.; Shrivastava, S.; Tzur, D.; Gautam, B.; Hassanali, M. DrugBank: A Knowledgebase for Drugs, Drug Actions and Drug Targets. Nucleic Acids Res. 2008, 36, D901–D906. [Google Scholar] [CrossRef]
  34. Butt, A.M.; Nasrullah, I.; Tahir, S.; Tong, Y. Comparative Genomics Analysis of Mycobacterium Ulcerans for the Identification of Putative Essential Genes and Therapeutic Candidates. PloS ONE 2012, 7, e43080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ferdous, S.; Akter, A. Identification of Potential Drug Targets by Subtractive Genome Analysis of Escherichia Coli O157: H7: An in Silico Approach. Adv. Appl. Bioinform. Chem. 2015, 8, 49–63. [Google Scholar]
  36. Yeh, I. Computational Analysis of Plasmodium Falciparum Metabolism: Organizing Genomic Information to Facilitate Drug Discovery. Genome Res. 2004, 14, 917–924. [Google Scholar] [CrossRef] [Green Version]
  37. Martínez, O.F.; Cardoso, M.H.; Ribeiro, S.M.; Franco, O.L. Recent Advances in Anti-Virulence Therapeutic Strategies with a Focus on Dismantling Bacterial Membrane Microdomains, Toxin Neutralization, Quorum-Sensing Interference and Biofilm Inhibition. Front. Cell Infect. Microbiol. 2019, 9, 74. [Google Scholar] [CrossRef] [Green Version]
  38. Topa, S.H.; Palombo, E.A.; Kingshott, P.; Blackall, L.L. Activity of Cinnamaldehyde on Quorum Sensing and Biofilm Susceptibility to Antibiotics in Pseudomonas Aeruginosa. Microorganisms 2020, 8, 455. [Google Scholar] [CrossRef] [Green Version]
  39. Butt, A.M.; Tahir, S.; Nasrullah, I.; Idrees, M.; Lu, J.; Tong, Y. Mycoplasma Genitalium: A Comparative Genomics Study of Metabolic Pathways for the Identification of Drug and Vaccine Targets. Infect. Genet. Evol. 2012, 12, 53–62. [Google Scholar] [CrossRef]
  40. Prosser, G.A.; Carvalho, L.P.S. De Kinetic Mechanism and Inhibition of Mycobacterium Tuberculosis D—Alanine: D—Alanine Ligase by the Antibiotic D—Cycloserine. FEBS J. 2013, 280, 1150–1166. [Google Scholar] [CrossRef]
  41. Bruning, J.B.; Murillo, A.C.; Chacon, O.; Barletta, R.G.; Sacchettini, J.C. Structure of the Mycobacterium Tuberculosis D-Alanine:D-Alanine Ligase, a Target of the Antituberculosis Drug D-Cycloserine. Antimicrob. Agents Chemother. 2011, 55, 291–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Howden, B.P.; Davies, J.K.; Johnson, P.D.R.; Stinear, T.P.; Grayson, M.L. Reduced Vancomycin Susceptibility in Staphylococcus Aureus, Including Vancomycin-Intermediate and Heterogeneous Vancomycin-Intermediate Strains: Resistance Mechanisms, Laboratory Detection, and Clinical Implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef] [Green Version]
  43. Kovač, A.; Konc, J.; Vehar, B.; Bostock, J.M.; Chopra, I.; Janežič, D.; Gobec, S. Discovery of New Inhibitors of D-Alanine:D-Alanine Ligase by Structure-Based Virtual Screening. J. Med. Chem. 2008, 51, 7442–7448. [Google Scholar] [CrossRef] [PubMed]
  44. Pandeya, A.; Ojo, I.; Alegun, O.; Wei, Y. Periplasmic Targets for the Development of Effective Antimicrobials against Gram-Negative Bacteria. ACS Infect. Dis. 2020, 6, 2337–2354. [Google Scholar] [CrossRef] [PubMed]
  45. Saiardi, A.; Azevedo, C.; Desfougères, Y.; Portela-Torres, P.; Wilson, M.S.C. Microbial Inositol Polyphosphate Metabolic Pathway as Drug Development Target. Adv. Biol. Regul. 2018, 67, 74–83. [Google Scholar] [CrossRef] [PubMed]
  46. Ramesh, P.; Nagendrappa, J.H.; Shivashankara, S.K.H. Comparative Analysis of Rosetta Stone Events in Klebsiella Pneumoniae and Streptococcus Pneumoniae for Drug Target Identification. Beni. Suef. Univ. J. Basic Appl. Sci. 2021, 10, 37. [Google Scholar] [CrossRef]
  47. Pirruccello, M.; Nandez, R.; Idevall-Hagren, O.; Alcazar-Roman, A.; Abriola, L.; Berwick, S.A.; Lucast, L.; Morel, D.; De Camilli, P. Identification of Inhibitors of Inositol 5-Phosphatases through Multiple Screening Strategies. ACS Chem. Biol. 2014, 9, 1359–1368. [Google Scholar] [CrossRef] [Green Version]
  48. Liao, J.; Wang, Q.; Wu, F.; Huang, Z. In Silico Methods for Identification of Potential Active Sites of Therapeutic Targets. Molecules 2022, 27, 7103. [Google Scholar] [CrossRef]
Figure 1. Complete workflow of the study.
Figure 1. Complete workflow of the study.
Biomedicines 11 02028 g001
Table 1. List of chokepoint enzymes from different pathways.
Table 1. List of chokepoint enzymes from different pathways.
Enzyme IDTotal No. of Pathways of the Enzyme
ECNA114_0085ena00473, ena00550, ena01100, ena01502
ECNA114_3261ena00550
ECNA114_1004, ECNA114_3778ena00540, ena01100
ECNA114_2045ena01053
ECNA114_0627ena00010, ena00030, ena00052, ena00230, ena00500, ena00520, ena00521, ena01100, ena01110, ena01120, ena01130
ECNA114_2137, ECNA114_2136ena00521, ena00523, ena01100, ena01130
ECNA114_2904, ECNA114_2317ena00071, ena00280, ena00310, ena00362,00380, ena00620, ena00630, ena00640,ena00650, ena00900, ena01100, ena01110, ena01120, ena01130, ena01200, ena01220, ena01212
ECNA114_1060ena00562, ena00627, ena01120
ECNA114_3742ena00910, ena01120, ena02020
ECNA114_1411ena00010, ena00071, ena00350, ena00625, ena00626, ena00650, ena01100, ena01110, ena01120, ena01130, ena01220
ECNA114_1698, ECNA114_1052, ECNA114_3080ena00633, ena01120
ECNA114_3742, ECNA114_3652ena00010, ena0071, ena00350,ena00625, ena00626, ena01100, ena01110, ena01120, ena01130, ena01150
ECNA114_4463ena02060, ena00500
ECNA114_2504ena00520, ena02060
ECNA114_1862ena00051, ena00520, ena01100, ena02060
ECNA114_4175, ECNA114_4173, ECNA114_4172, ECNA114_2735, ECNA114_3748, ECNA114_2977ena00051, ena02060
ECNA114_3218, ECNA114_3216, ECNA114_3217, ECNA114_3224,ena00052, ena02060
Table 2. List of virulence genes.
Table 2. List of virulence genes.
Virulence Factors UPECGenes NameFound in UPEC Strain NA114 and Not in Human
Iron uptakeiutA, iucA, iucB, iucC, iucDiucA, iucB, iucD, iucC
Chu (E. coli hemin uptake)chuA, chuS, chuT, chuU, chuV, chuW, chuX, and chuYchuU, chuW
EnterobactinentA, entB, entC, entD, entE, entF, fepA, fepB, fepC, fepD, fepE, and fepGfepA, fepB, fepC, fepD, fepG, entE, entA, entB, entF, entC
IroNiroNiroN
HemolysinhlyA, hlyB, hlyC, and hlyDhlyB, hlyD
Table 3. List of selected resistance genes.
Table 3. List of selected resistance genes.
Resistance Gene Found in String after ARDB ToolFound UPEC Strain NA114Found in HumanEnzyme No.
acrBYesNoc0580
acrAYesNoc0581
macBYesNoc1016
arnAYesNoc2797
tolCYesNoc3781
bacAYesNoc3807
Table 4. List of selected essential genes.
Table 4. List of selected essential genes.
Sr. No.Query ProteinNo. of a Homolog in DEGDEG Accession Number
1ECNA114_00851DEG10180021
2ECNA114_10042DEG10190079, DEG10180150
3ECNA114_37783DEG10480267,DEG10180536, DEG10190246
4ECNA114_20451DEG10180357
5ECNA114_21371DEG10180210
6ECNA114_36521DEG10180338
7ECNA114_10521DEG10180159
8ECNA114_18621DEG10180464
9ECNA114_32181DEG10180032
10entD1DEG10190060
11entE1DEG10180357
12fepB1DEG10180106
13fepC2DEG10190239; DEG10480100
14arnAc27973DEG10180489, DEG10480227, DEG10190203
15ECNA114_05801DEG10480311
Table 5. Predicted subcellular locations of putative drug targets.
Table 5. Predicted subcellular locations of putative drug targets.
Protein Enzyme CodeCelloPSORTbProtCompbSubcellular Location
ECNA114_4463InnermembraneCytoplasmic MembraneInnermembraneInnermembrane
ECNA114_4172InnermembraneCytoplasmic membraneInnermembraneInnermembrane
ECNA114_2735InnermembraneCytoplasmic membraneInnermembraneInnermembrane
ECNA114_3216InnermembraneCytoplasmic membraneInnermembraneInnermembrane
ECNA114_3224InnermembraneCytoplasmic membraneInnermembraneInnermembrane
ECNA114_0085CytoplasmicCytoplasmicCytoplasmicCytoplasmic
ECNA114_1060PeriplasmicCytoplasmicPeriplasmicPeriplasmic
Table 6. List of identified putative drug targets.
Table 6. List of identified putative drug targets.
Putative Targets ID’sFunctionPathways InvolvedDruggability AnalysisSubcellular Location
ECNA114_4463treB, PTS system,Phosphotransferase system,
Starch and sucrose metabolism
NoInnermembrane
ECNA114_4172PTS systemMannose and fructose metabolism,
Metabolic pathways, Phosphotransferase system
NoInnermembrane
ECNA114_2735srlE, PTS systemMannose and fructose metabolism,
Metabolic pathways, Phosphotransferase system
NoInnermembrane
ECNA114_3216agaW, component of PTS systemGalactose metabolism, Metabolic pathways, Phosphotransferase systemNoInnermembrane
ECNA114_3224agaD, a component of the PTS systemGalactose metabolism, Metabolic pathways, Phosphotransferase systemNoInnermembrane
ECNA114_0085D-alanine-D-alanine ligaseD-alanine metabolism, Metabolic pathways, Vancomycin resistance, Peptidoglycan biosynthesisYesCytoplasmic
ECNA114_1060appA, Phosphoanhydride phosphohydrolaseInositol phosphate and riboflavin metabolism, Metabolic pathwaysNoPeriplasmic
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

Kaur, H.; Modgil, V.; Chaudhary, N.; Mohan, B.; Taneja, N. Computational Guided Drug Targets Identification against Extended-Spectrum Beta-Lactamase-Producing Multi-Drug Resistant Uropathogenic Escherichia coli. Biomedicines 2023, 11, 2028. https://doi.org/10.3390/biomedicines11072028

AMA Style

Kaur H, Modgil V, Chaudhary N, Mohan B, Taneja N. Computational Guided Drug Targets Identification against Extended-Spectrum Beta-Lactamase-Producing Multi-Drug Resistant Uropathogenic Escherichia coli. Biomedicines. 2023; 11(7):2028. https://doi.org/10.3390/biomedicines11072028

Chicago/Turabian Style

Kaur, Harpreet, Vinay Modgil, Naveen Chaudhary, Balvinder Mohan, and Neelam Taneja. 2023. "Computational Guided Drug Targets Identification against Extended-Spectrum Beta-Lactamase-Producing Multi-Drug Resistant Uropathogenic Escherichia coli" Biomedicines 11, no. 7: 2028. https://doi.org/10.3390/biomedicines11072028

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