Open Access

Molecular mechanisms of multidrug resistance in clinically relevant enteropathogenic bacteria (Review)

  • Authors:
    • Julia Alexsandra González-Villarreal
    • Katia Jamileth González-Lozano
    • Elva Teresa Aréchiga-Carvajal
    • Jesús Antonio Morlett-Chávez
    • Miriam Paulina Luévanos-Escareño
    • Nagamani Balagurusamy
    • Mauricio Andrés Salinas-Santander
  • View Affiliations

  • Published online on: November 8, 2022     https://doi.org/10.3892/etm.2022.11689
  • Article Number: 753
  • Copyright: © González-Villarreal et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Multidrug resistant (MDR) enteropathogenic bacteria are a growing problem within the clinical environment due to their acquired tolerance to a wide range of antibiotics, thus causing severe illnesses and a tremendous economic impact in the healthcare sector. Due to its difficult treatment, knowledge and understanding of the molecular mechanisms that confer this resistance are needed. The aim of the present review is to describe the mechanisms of antibiotic resistance from a genomic perspective observed in bacteria, including naturally acquired resistance. The present review also discusses common pharmacological and alternative treatments used in cases of infection caused by MDR bacteria, thus covering necessary information for the development of novel antimicrobials and adjuvant molecules inhibiting bacterial proliferation.

1. Introduction

Gastrointestinal diseases are the most frequent cause for medical consultation and one of the leading causes of death worldwide (1,2). In America, 77 million individuals get sick annually due to food poisoning and, according to the World Health Organization (WHO), one in ten individuals get sick each year for the same reason worldwide. As a result, ~420,000 individuals die on a yearly basis, of which ~30% are children under five years of age. It must be noted that diarrheal diseases correspond to more than half of the cases of gastrointestinal illnesses, for which ~95% of cases can be associated with Campylobacter spp., Escherichia coli and non-typhoidal Salmonella spp. (3).

In 2017, the WHO published a list of drug resistant bacteria for which there is a growing need to develop new antibiotics as even the current most effective of them, such as carbapenems and cephalosporins, are now ineffective. This list is divided into three categories (critical, high, and medium priority) based on how urgently these antibiotics are needed. The critical priority group includes multidrug resistant (MDR) bacteria that are especially dangerous for vulnerable individuals, or individuals under specialized care, due to the high risk of infection, complications, disease severity and mortality (4). Some of the bacteria included in this group are: Acinetobacter spp., Pseudomonas spp., Klebsiella spp., Escherichia coli, Serratia spp. and Proteus spp. (4), all of which have different infection pathways in the host (5).

Different molecular mechanisms of bacterial resistance to antibiotics have been described so far. Due to the importance of this phenomenon in public health, the present review gathers engaging and relevant information concerning the most common enteropathogenic bacteria in clinical practice and describes the molecular mechanisms for the acquisition or de novo development of antibiotic resistance, thus seeking to enlighten the reader in this regard and provide a greater understanding of this process.

Thorough research was conducted in the writing of the present manuscript, primarily employing informatic tools such as PubMed (https://pubmed.ncbi.nlm.nih.gov/), Scopus (https://www.scopus.com/home.uri), Scielo (https://scielo.org/), Medigraphic (https://www.medigraphic.com/newMedi/) and Science Direct (https://www.sciencedirect.com/). The terms used in this search included: Enterobacteria, multidrug resistance, enteropathogenic bacteria, multidrug-resistant bacteria, bacterial drug resistance, horizontal gene transfer and gastrointestinal diseases. Inclusion criteria included English language and full-length articles. Exclusion criteria: Publications from 2012 to 2022 were prioritized. Older publications were also reviewed and introduced in the present study if deemed relevant. A total of 99 research and review articles were used in the present study (Fig. 1).

2. Multidrug resistant enteropathogenic bacteria

The family Enterobacteriaceae includes several genus and species of both gram-negative and -positive bacilli (for example Enterococcus spp.), a number of which are present in water, soil, plants and the intestinal microbiota of humans and animals; however, their diversity is often dictated by geographical area (6) and often develop as opportunistic pathogens causing severe infections in humans (Table I) (7).

Table I

Common enteropathogenic bacteria in clinical practice and their symptoms.

Table I

Common enteropathogenic bacteria in clinical practice and their symptoms.

BacteriaAntibiotic resistancePathologyMechanism of action(Refs.)
Klebsiella pneumoniaeCarbapenems, β-lactams, aminoglycosides, quinolones, tigecycline, polymyxin.Acute Diarrheic Syndrome, urinary tract infections, cystitis, pneumonia, endocarditis, septicemia.Adherence and biofilm formation by type 1 and type 3 pili.(8,85,86)
Escherichia coliCephalosporins, fluoroquinolones.Acute watery diarrhea, bloody diarrhea.A/E and changes of the host apical enterocyte membrane. Activation of T3SS and formation of A/E.(2,5,8,87)
Shigella spp.Cephalosporins, ampicillin, co-trimoxazole, nalidixic acid.Bloody diarrhea.T3SS encoded on a large plasmid and transport of effector proteins.(5,10,88,89)
Salmonella spp.Quinolones, nalidixic acid.Acute watery diarrhea, bloody diarrhea, enteric fever.Activation of T3SS and transport of effector proteins.(5,10,90)
Campylobacter spp.Quinolones, tetracycline.Enteric fever, acute watery diarrhea, bloody diarrhea.Presence of flagella, high molecular weight plasmids, surface adhesins and chemotactic factors.(10,90,91)
Vibrio choleraeAmpicillin, nalidixic acid, co-trimoxazole.Acute liquid diarrhea.Biofilms formation and production of extended-spectrum β-lactamases.(90,92,93)
Aeromonas spp.Beta-lactams, tetracyclines, glycylcyclines, fluoroquinolones, aminoglycosides, sulfamethoxazole-trimethoprim.Acute watery diarrhea, bloody diarrhea.Travel by the blood to the first organ it finds where it produces the toxic enterotoxin aerolysin.(2,94,95)
Yersinia enterocoliticaNalidixic acid.Enteric fever, bloody diarrhea.Activation of T3SS and transport of effector proteins and/or apoptosis. Yersinia forms microcolonies and starts replication.(5,10,90,96)
Staphylococcus aureusPenicillin, methicillin.Acute liquid diarrhea.Inoculation into an open wound. adhesion and invasion of host epithelial cells by microbial surface components recognizing adhesive matrix molecules.(2,90,97)
Enterococcus spp.Vancomycin, Beta-lactams, glycopeptides, aminoglycosides, tetracyclines, quinolones, macrolides.Sepsis, endocarditis, urinary tract infections.When pathologic alterations are caused by either direct toxin activity or indirectly by bystander damage from the inflammatory response, enterococci are able to outpace host defenses, multiply at rates that are faster than clearance, and overwhelm the host.(18,98,99)

[i] A/E, attaching/effacing lesion; T3SS, Type III secretion system.

These bacteria are associated with 10-20% cases of infectious diarrhea in children worldwide (8,9). The majority of patients affected by these bacteria only require an hydroelectrolytic imbalance intervention, caused by dehydration or antibiotics treatment, the latter of which diminishes the duration of the disease, reduces its transmission and prevents complications (10). In some cases, it is possible that severe infections can be caused by multidrug-resistant enterobacteria or by enterotoxin producing bacteria, and for this reason special epidemiological surveillance is necessary (10,11). A report made in 2017 revealed that antibiotic resistance in Latin America was as high as 45%, followed by Europe with 39%, the US with 8%, and Canada with 5% (12).

In 2019 Levin-Reisman et al (13) described the different phenotypic traits enabling bacteria to acquire resistance to antibacterial agents, such as tolerance, persistence and resistance. Tolerance is the ability of a bacterial population to survive and grow under toxic conditions, such as high concentrations of antibiotics, thus prolonging treatment duration; notably, this acquired resistance may or may not be inherited to daughter cells (13-15). Persistence is the ability of bacteria to survive a specific drug concentration, prolonging the duration of treatment unless corrected (13). These persistent bacteria can withstand antibiotic treatment without affecting the drugs' minimum inhibitory concentration (MIC), presenting a biphasic death curve because the majority of the bacterial population dies, with only a small subpopulation persisting for a longer time (13-15). Resistance is the ability to grow in the presence of environmental stress or high concentrations of antibiotics, regardless of the treatment's duration, due to the increased MIC required to effectively destroy the microorganism (13-16).

The acquisition of antibiotic or antimicrobial resistance is a natural selection process of bacteria and thus considered as part of their evolutionary path. In this regard, the indiscriminate use of antibiotics exerts a high selective pressure on them, which results in genomic changes that translate into multidrug resistance, as seen with greater frequency in developing countries (15,17).

Depending on the number or type of antimicrobials, resistant bacteria can be classified as MDR, which occurs when clinically relevant microorganisms have developed resistance to three or more classes of commonly used antibiotics and/or antimicrobials (18,19); extensively drug-resistant (XDR), microorganisms resistant to at least one agent of all antimicrobial classes; and pandrug-resistant, which includes microorganisms resistant to all agents in all antimicrobial classes (20). Most of these multidrug-resistant bacteria are typically gram-negative enterobacteria representing an important therapeutic challenge in the treatment of life-threatening infections (12,15).

3. Molecular mechanisms of multidrug resistance

Antibiotic resistance can be permanently maintained once it has been fixed in the genome or it can be just temporary if the selective pressure is absent causing non-resistant bacteria to proliferate instead. Drug resistance often appears due to the acquisition of exogenous DNA or through genomic DNA mutations (21).

From an evolutionary perspective, bacteria have several advantages over other organisms because they have short replication time, large populations and capacity for horizontal gene transfer, which enables bacteria to adopt, use, propagate and fix advantageous genetic information between strains and species, such as antibiotic resistance (22).

The limitation of both resources and nutrients within the environment is a decisive factor exerting great selective pressure on bacteria, forcing the stressed populations to adapt or die. As a result, the genetic variations providing a survival advantage become fixed in the bacterial population, thus taking another step in their evolution as a species (23,24).

The genetic evolution of bacteria mostly occurs due to recombination events allowing gene acquisition, segment duplication, fusion of homologous regions, functional domain exchange and gene deletion (24,25). Acquisition of exogenous genomic material occurs via horizontal gene transfer (HGT), which enables bacteria to absorb and incorporate genetic material of diverse origin, thus giving rise to different genotypes between populations of the same species. Further, HGT events can also confer pathogenicity factors related to virulence, symbiosis, resistance and metabolism, among others (24,25).

Three major mechanisms of HGT have been described until 2019: i) Natural transformation (26); ii) conjugation (27); and iii) transduction (28). However, Soler and Forterre (25) proposed a fourth mechanism called vesiduction in 2020 (Fig. 2).

Natural transformation

This phenomenon represents the active transfer of genes from extracellular free DNA from lysed bacteria to a living, competent bacterium that captures and incorporates it into its genome through DNA recombination. This process contributes to genetic variability, shapes evolution and, in the case of pathogenic bacteria, it is responsible for their adaptation to host cells, fosters the spread of antibiotics resistance, promotes antigenic variation and leads to the acquisition of new virulence factors (29-31). In addition, natural transformation also promotes DNA exchange between taxonomically distant bacteria (29-31).

In 2018, Ellison et al (31) demonstrated the ability of bacteria to capture and introduce free DNA molecules through surface appendages known as competing pili. Using Vibrio cholerae as a model, type IV competing pili were demonstrated to be able to capture and bind double-stranded DNA from the extracellular space. Once bound to DNA, the pili retracts and mobilizes the captured DNA molecule towards the cell surface, where it is finally internalized.

Conjugation

This process explains the exchange of genetic material from donor bacteria with an adjacent recipient through a sexual pilus or physical contact, requiring the formation of a pore between both bacteria while connected as a mating pair. The exchanged genetic elements can usually provide resistance to drugs, antiseptics and/or disinfectants (30,32).

The occurrence of a conjugation event, and thus of an effective DNA transfer, requires cell-cell contact between a donor and a recipient cell. There are two types of genetic elements than can be exchanged during this conjugation: i) Conjugative plasmids, found in free form within the intracellular space and with autonomous replication capacity; and ii) integrated-conjugative elements, or conjugative transposons, that can integrate into the genome of the recipient cell. Since these plasmids were part of the donor's genome prior to the exchange, the latter are rarely, if ever, found free in the cytoplasm and do not replicate autonomously (33,34). In the majority of bacteria, conjugation occurs by transferring single-stranded DNA molecules through the type IV secretion system contained in the conjugative element, which can also transfer DNA from bacteria to eukaryotes (33,34).

Transduction

Transduction is mediated by bacterial viruses called bacteriophages. When a phage infection culminates in bacterial lysis, some viral particles can encapsulate bacterial DNA fragments, thus producing transducer particles. Upon subsequent infection, the transducer particles inject bacterial DNA into the next bacterium host, which may acquire new genetic traits after adopting the exogenous DNA (35). Generalized transduction occurs when any of the bacterial genes maintains the same probability of being encapsulated in a transducing particle and transferred into a recipient. On the other hand, specialized transduction defines the transference of specific genes, such as those located next to the bacteriophage's DNA (30,36,37).

Vesiduction

Vesiduction was proposed in 2020 by Soler and Forterre (25). It involves the donation and/or acquisition of exogenous material from extracellular vesicles, a phenomenon that has been observed in all three domains of life. Vesicles are secreted through the cell membrane of Gram-positive bacteria and the outer cell membrane of Gram-negative bacteria. The precise mechanism is not yet fully understood, and it may be possible that it differs according to the composition of the cell wall and the proteins used in the construction of the vesicles (38,39). These vesicles can fulfill different physiological roles that are not mutually exclusive with genetic material transference, such as the transport of peptidoglycan hydrolases or toxins, and other effector proteins that may be involved in the elimination of concurrent microorganisms through competition or pathogenicity. Some vesicles can also transport intercellular communication molecules (39).

Regardless of their inherent differences, all of the previously mentioned mechanisms for genetic acquisition can be driven by RecA-dependent recombination, illegitimate recombination, transposition or integration (25).

4. Molecular mechanisms of multidrug resistance generation

There are different mechanisms of natural resistance that can appear through other pathways; however, these are usually induced by the presence or prolonged exposure of hazardous molecules (such as antibiotics) resulting in the proliferation of those populations with advantageous biological changes (40,41). The majority of these mechanisms are specifically developed by bacteria to generate resistance to antibiotics or antimicrobials and may involve the modification of existing genomic material through spontaneous mutations that might be punctual or massive. These resistant populations thrive thanks to the action of bactericidal molecules eliminating the cells lacking tolerance or resistance; in other words, the microorganisms are forced to evolve in order to survive (21). This selective pressure has become the standard in areas such as hospitals, biohazard waste disposal areas, pharmaceutical industry effluents, wastewater, manure treated soils, animal breeding and aquaculture areas (21).

Inherent (natural) resistance

Natural resistance to drugs, antibiotics or antimicrobials is a trait often shared between different species of microorganisms, which may be due to the same physiology or spontaneous genetic mutations regardless of previous exposure to these molecules (42,43). An example of intrinsic antibiotics resistance conferred by physiology can be seen in bacteria of the Mycoplasma genus, whose members are highly resistant to drugs targeting the cell wall, such as β-lactams and glycopeptides (44,45); although some antibiotics normally have difficulty crossing the outer membrane of Gram-negative bacteria. For example, vancomycin inhibits cell wall synthesis by targeting d-Ala-d-Ala peptide precursor units of Gram-positive bacteria, thus preventing the assembly of peptidoglycan layers and transpeptidation (46). By contrast, this antibiotic cannot affect Gram-negative bacteria since it is unable to cross the outer membrane, and thus kept from accessing the cell wall (46). Even though these events occur naturally in the environment, (47) it must be mentioned that the intrinsic resistance to antibiotics is not considered as a clinical problem because previously developed antibiotics do not target these bacteria.

Spontaneous mutations

Spontaneous mutations occur by random nucleotide changes that induce different effects; for example, amino acid sequence variations that may lead to altered phenotypes. These mutations can be caused by DNA replication errors or through the action of mutagenic agents. It must be noted that acquired mutations are often detrimental, so these are usually not inherited, are rarely widespread and often are just isolated events (21,48). However, when a mutation provides a biological advantage, this change can become fixed in the population through vertical gene transfer and become a dominant trait (21,48). The frequency of spontaneous mutations related to antibiotic resistance occur at a rate of 1x10-5 to 2x10-8 in members of the Chlamydiaceae and Helicobacter pylori (49,50). Though this would appear to be a rare event, in reality antibiotic resistance appears in bacterial populations within a relatively short period of time, accelerating further when exposed to a selective agent due to exponential growth rate and the number of cells generated per replication cycle (51). For example, the gastric pathogen Helicobacter pylori can have different mutations in the 23S rRNA, gyrA and rpoB genes, which are responsible for resistance to clarithromycin, ciprofloxacin and rifampicin, respectively (50). The capacity of Chlamydia trachomatis to resist antibiotics such as azithromycin, tetracycline and fluoroquinolone has also been attributed to spontaneous mutations (52). Although this mutation rate is not even across the board, there are bacterial subpopulations with a significant tendency to acquire and accumulate spontaneous mutations, which is why they often present a greater number of mutation events compared with what is commonly observed (21). These subpopulations are known as hypermutable and, although not all spontaneous mutations confer antibiotics resistance, this hypermutability is directly proportional with the increased resistance capacity (21).

Duplications

Gene duplications are often overlooked as the primary source of functional genomic diversity, originating new functions from a pre-existing gene. In addition, the generation of genetic copies derives into elements that can evolve independently due to inexistent selective pressure, further diversifying their functions (53). For example, the Plasmodium falciparum multidrug resistance protein transporter 1 gene plays an important role in the parasite's resistance to drugs due to the strong correlation between the number of copies and the resistance to artemisinin-based therapies, an anti-malaria drug used to reduce its mortality rate since the year 2000. By 2020, Calçada et al (54) reported the threat of resistance against this drug due to the appearance of new duplication events and the presence of single nucleotide polymorphisms in current strains.

5. Drugs and bacterial response

As aforementioned, the genetic elements leading to drug resistance can be spread between different microorganisms in different manner, from the horizontal (transformation, transduction, conjugation or vesiduction) to vertical gene transfer (from mother to daughter cells) of either intrinsic or extrinsically acquired genomic modifications, such as spontaneous mutations, duplications, insertions, deletions or transpositions. The mechanism of antibiotics resistance is highly dependent on the way the drug itself works against the bacterium, regardless of how this resistance was acquired, thus deriving in different survival pathways that may limit the absorption of drugs, modify the target molecules, directly inactivate the drug and/or secrete it into the microenvironment (Fig. 3) (55).

6. Mechanisms of antibiotics

Antibiotics with the capacity to inhibit or kill a wide range of bacteria are known as broad-spectrum antibiotics, whereas that those that only affect certain types of bacteria are known as narrow-spectrum antibiotics. Antibiotics typically target the structure or metabolic processes of bacteria, preventing their replication (56-58). In this regard, the most common mechanisms consist in the inhibition of cell wall synthesis, DNA replication or transcription, protein synthesis, metabolic pathways or directly through cell membrane degradation (41,57).

Cell wall synthesis inhibition

The majority of bacterial cells are surrounded by a rigid peptidoglycan layer consisting of long sugar polymers linked through peptide bonds. This structure is needed for survival as it protects the bacteria from osmotic pressure and other hostile conditions from the environment (56,57,59). Drugs such as penicillin and cephalosporins inhibit the formation of peptide bonds in the bacterial cell wall, thus effectively killing the microorganism (56). By contrast, glycopeptides inhibit bacterial growth by inhibiting peptidoglycan synthesis (56,57).

Cell membrane function inhibition

In comparison with gram-positive bacteria, gram-negatives have a greater resistance to antimicrobials due to the existence of an external cell membrane regulating both intracellular and extracellular substance flow (56,60). The drugs targeting this external cell membrane are specific for each microbial group because their function depends on the lipid content of such membrane; however, these drugs can sometimes be toxic, thus limiting their use (56,57). For example, Daptomycin can rupture the cell membrane due to depolarization, whereas that polymyxins bind to the lipid fraction of the membrane's lipopolysaccharide layer, thus causing its disintegration (57).

Nucleic acid synthesis inhibition

Nucleic acid synthesis is important for the survival of living beings, including bacteria. The cellular processes responsible for cell replication and bacterial conservation can be negatively affected due to the interruption of this process by drugs that block DNA replication or transcription (56,57). In this regard, antibiotics such as quinolones interfere with the functionality of the helicase enzyme preventing the function of unwinding DNA, effecting the process of DNA replication and repair. On the other hand, they can exert their action by inhibiting topoisomerase II and IV of bacteria, preventing the synthesis of RNA (61).

Metabolic inhibitors

Some drugs act against important metabolic processes for survival, such as the folic acid pathway, which is necessary to produce important precursors in DNA synthesis (57). In this case, sulphonamides and trimethoprim release similar substrates to those produced and used by the bacteria in its normal metabolism (56,57). Each of these drugs is responsible for inhibiting different stages of folic acid metabolism. For example, the sulphonamides competitively inhibit dihydropteroate synthase, binding to it with greater affinity compared with the substrate produced by the bacteria; while trimethoprim is responsible for inhibiting dihydrofolate reductase at a later stage of folic acid synthesis (56,57,59).

Protein synthesis inhibition

Proteins play a role in various cellular structures and physiological processes; therefore, their synthesis is fundamental for survival (56,57). For this reason, drugs that inhibit protein biosynthesis by targeting the 70S prokaryotic ribosome (30S and 50S ribosomal subunits) constitute the largest class of antibiotics (56,57,59).

30S subunit inhibitors

Antibiotics such as tetracycline, aminoglycosides and streptomycin target and inhibit the 30S ribosome, blocking the passage of aminoacyl-tRNA towards the ribosome (57,59).

50S subunit inhibitors

Antibiotics targeting the 50S ribosomal subunit can act in two different ways, by blocking protein translation (oxazolidinones) or by blocking the elongation phase of protein synthesis (for example, macrolides). However, the latter may be ineffective when the elongation phase has advanced significantly (57,59). Natural antibiotics such as aminoglycosides are considered as bactericidal, whereas macrolides, tetracyclines, chloramphenicol, streptogramins and spectinomycin are considered as bacteriostatic (57).

7. Mechanisms of drug resistance

Antibiotics outlet, secretion or efflux pumps

Some bacteria have exporter proteins on their cell membrane that can rapidly transport the antibiotics from the cytoplasmic membrane in gram-positive bacteria and from the intermembrane space in gram-negative bacteria to the exterior of the cell without the help of energy-dependent efflux pumps, thus preventing the antibiotics from reaching their target (30,62,63). There are two groups of efflux pumps, some of them are specific whereas others can secrete diverse substances. These pumps are classified according to energy source and function; in this regard, the first group uses ATP as an energy source and functions through hydrolysis (63,64). By contrast, the second group uses the mobile force of protons as an energy source, enhancing secretion through the electrochemical potential of the membrane (63,64). A total of five families of efflux pumps have been described within the second group: i) The multidrug and toxic extrusion family; ii) the major facilitator super family; iii) the resistance nodulations cell division (RND) family; iv) the small MDR family; and v) the multidrug endosomal transporter family (62-64). These efflux pumps are widely distributed among gram-positive and -negative bacteria, except for the RND poly-selective superfamily, which is found gram-negative bacteria with very high frequency (62,63). These efflux pumps play a notable role in multidrug resistance due to their capacity to secrete a wide range of structurally unrelated drugs and molecules (62-64).

Permeability alterations in the outer cell membrane

The majority of antibiotics penetrate the bacterial membrane and target diverse intracellular processes; therefore, the concentration of antibiotics within the cell can be affected by alterations in the lipid bilayer of the membrane, modifying either the cell's diameter or number of porins (30,65). The bacterial cell envelope provides a selective barrier allowing the exchange of nutrients and signaling molecules with the microenvironment. This envelope is formed by the cell wall and the plasma membrane, and, in Gram-negatives, it provides an additional function as a physical barrier that reduces the permeability of a number of drugs (53,65). Notably, this envelope can also be targeted by antibiotics (53,65). The outer membrane of gram-negative bacteria is populated by proteins called porins, which determine its permeability and allow the entry of hydrophilic compounds into the cell. The absence or low number of porins can also prevent the entry of antimicrobial molecules, thus hindering their action in the cytoplasm and/or cell envelope (62,63).

Active site alterations

Bacteria have the ability to form metabolic substances that compete with antimicrobial drugs for the active site, preventing it from binding due to loss of affinity (30,63). There are two types of modifications in this regard as follows.

Penicillin-Binding-Protein (PBP) modification. Observed in Gram-positive bacteria, this effect is caused by variations in the peptidoglycan gene, which modify the antimicrobial binding site in the cell wall (30,62).

Ribosomal modification. The ermA and ermB genes can modify the ribosome's active site through methylation. These modifications occur in the 30S and 50S subunits of the 70S ribosome, affecting the target site of drugs such as aminoglycosides, macrolides, tetracyclines and lincosamides (30,66).

Enzymatic modification or inactivation of antibiotics

This is the most common mechanism of resistance observed in bacteria. It is achieved through the expression of enzymes with the capacity to modify the active component of the antibiotics, thus reducing their effectiveness. Three mechanisms have been reported so far: i) Redox reactions; ii) group transfer; and iii) enzymatic hydrolysis. The latter is the primary mechanism of resistance, with the clearest example being the hydrolysis of the beta-lactam ring of antibiotics. The enzymes of gram-negative bacteria typically originate from a plasmid or have a transposon origin with constitutive and periplasmic expression. By contrast, this resistance is solely provided by a plasmid in gram-positive bacteria, which can be inducible and/or extracellular (40,65).

Biofilm formation

Biofilms are structured aggregations of bacterial cells enclosed in a self-synthesized extracellular matrix composed of different macromolecules such as proteins, nucleic acids and polysaccharides (63,67). Biofilms bacterial production protects them from ultraviolet light, dehydration, immune system or certain antibiotics. There are three important steps in biofilm formation: i) Adhesion, in this phase bacteria can attach to any give surface; ii) growth and maturation, occurs when bacteria secrete an exopolysaccharides matrix and mature from microcolonies to multi-layered cell clusters; and iii) shedding, which can be either active (initiated by the bacteria) or passive (caused by external factors) (30,62). Amongst the most common pathogens that develop biofilms are S. aureus, P. aeruginosa, A. baumannii and K. pneumonia (30,62).

Target site overexpression

This mechanism has been described in clinical isolates of mycobacteria with promoter duplications. This often results in the overexpression of genes that may include mutations affecting the target site of antibiotics or antimicrobials (30). In this regard, Martinez et al (68) describe the presence of plasmids in E. coli that provide resistance to amoxicillin-clavulanate as a result of the hyperproduction of plasmid-determined TEM-1 P-lactamase. TEM-1 β-lactamase is a known determinant of resistance to antibiotics, such as penicillin, cephalosporins and their derivatives, including second, third and fourth generation cephalosporins, monobactams and β-lactamase inhibitors. This enzyme inactivates the aforementioned compounds by hydrolyzing their lactam rings (69,70).

8. Treatment against multidrug-resistant bacteria

Some of the first-line drugs used in the treatment of serious infections caused by Enterobacteriaceae include penicillin, cephalosporins, carbapenems, fluoroquinolones, monobactams and, occasionally, aminoglycosides. However, bacterial resistance against these drugs is rapidly becoming widespread, thus making difficult these treatment (20,71). In some cases, second-line drugs are more effective against enterobacteria, as would be the case with polymyxins, tigecycline, aminoglycosides and fosfomycin (72). Pathogenic bacteria have evolved different strategies to overcome the host's response by avoiding highly competitive environments. For example, the mucosal barrier can be breached by mucinases, such as the Pic enzyme from Shigella and enteroaggregative from Escherichia coli (EAEC). Notably, the Pic gene can be found in a ‘pathogenicity island’ flanked by insert-like EAEC elements that have been acquired through horizontal gene transfer (24).

Empirical treatment with antibiotics

As a first line decision, empirical therapy becomes essential in the treatment of serious infections caused by bacteria. However, the emergence of bacterial resistance complicates its implementation, thus causing a serious dilemma between the selection of a broad-spectrum drug, which could induce greater drug resistance, or a narrow-spectrum drug, which could be completely ineffective (71,73). Regardless of its potential shortcomings, the latter could supply important information on the pathogen's susceptibility to certain antimicrobials (71,73). Several factors must be evaluated during the selection of antibiotics treatment, including susceptibility, risk of developing resistance, potential side effects, comorbidities, local epidemiology and clinical severity (10,71,74).

Combination antibiotic therapy, The combined therapy of antibiotics enables the synergistic effect of one or more drugs, potentially increasing the probability of an effective treatment and lowering the risk of bacterial resistance. However, the results of drug synergy tests observed in vitro do not always translate well into a clinical setting (71,75).

Alternative treatments

Alternative treatments can also be implemented in addition to antibiotics therapy if their contribution proves safe for the patient and does not enable the development of bacterial resistance, for example, phage therapy or competing microorganisms (76). There are some reports demonstrating the benefits of these treatments against multidrug-resistant pathogens, even suggesting they could be used as replacements for common drugs (76).

Phage therapy

Bacteriophages are bacteria-specific viruses that can infect bacteria through the binding of specific receptors on the cell's surface and injecting their genetic material. Once infected, the phage can go through a lysogenic cycle, where the phage's genome is integrated in the bacterial chromosome as an endogenous prophage, spreading horizontally during cell division. The virus can remain latent for prolonged periods of time during this cycle; however, environmental or cellular stress factors can re-activate the phage and induce its lytic cycle, in which the viral genome is no longer integrated in the bacterial chromosome and goes into a massive replication event, finally causing cell death after the phage's lytic proteins hydrolyze the cell wall. These liberated phage particles can then infect other bacteria and start the lytic cycle again (76-78). It must be mentioned that the infective capacity of bacteriophages is constrained to particular bacterial species, thus resulting in a reduced spectrum. Although this could be considered a shortcoming, it could also be considered as a positive aspect since they are unable to affect the intestinal microbiota or the host (76,78).

Probiotics, prebiotics and synbiotics

Probiotics are live microorganisms that play a beneficial role if administered in adequate amounts, regardless of those present in the essential diet or naturally in the intestinal microbiota of the host (79-81). On the other hand, prebiotics are non-digestible compounds (non-starch polysaccharides and non-digestible oligosaccharides) present in the daily diet and which help stimulate the growth or activity of the intestinal microbiota, favoring the development of beneficial microorganisms (79,80,82). Finally, synbiotics are a composition of the previous two and which are often found in the form of pharmaceutical or food preparations containing one or more probiotic organisms and prebiotic compounds in order to provide a synergistic effect on the prebiotics, enhancing the development, activity and nutritional properties of the probiotics. The inclusion of synbiotics increases the density of probiotics and their health benefits (80,82). The probiotics used in clinical treatments are mainly composed of Gram-positive strains such as Lactobacillus that are resistant to the human digestive process. The administration of these microorganisms improves the epithelial barrier function, promotes the growth of beneficial bacteria, the proliferation of epithelial cells in the host (by upregulation of cell growth and downregulation of apoptosis), prevents the adhesion and colonization of pathogenic microorganisms and toxins, improves lactose digestion, produces antimicrobial peptides, regulates the immune response and improves the ability to regulate pH (76,83). Regarding the functional foods that seem to exert the best prebiotic effect, fructooligosaccharides, galacto-oligosaccharides and xyloseoligosaccharide, inulin and lactulose, can be mentioned. Some extracted from sources such as chicory and yacon roots, are reported (84). These can be used in symbiotic formulations with Lactobacilli, Bifidobacteria spp, S. boulardii and B. coagulans, among other probiotic agents (84).

9. Conclusions

Several clinically relevant bacterial strains are now resistant to multiple drugs. To counteract this phenomenon, novel compounds with the capacity to kill and/or prevent their proliferation are constantly being developed. However, the epidemiology and resistance of these strains varies widely according to geographical region. Therefore, alternative treatments are also being sought to enhance the effectiveness of antibiotics to reduce bacterial proliferation and prevent further spread of antibiotics resistance.

Acknowledgements

Thanks to Dr Daniel Díaz (Department of Pharmacology, Faculty of Medicine in Hradec Králové; Charles University, Prague, Czech Republic) for his kind assistance in proofreading and editing this manuscript.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JAGV, KJGL and MASS contributed to the study design, and performed the literature review and data collection. JAGV, KJGL, ETAC and MASS contributed to the selection the relevant literature and critical interpretation. JAGV, KJGL, ETAC, MASS, JAMC, MPLE and NB drafted and improved the manuscript. JAGV, KJGL, ETAC, MASS, JAMC, MPLE and NB critically read and modified the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sell J and Dolan B: Common gastrointestinal infections. Prim Care. 45:519–532. 2018.

2 

Hernández CC, Aguilera AMG and Castro EG: Gastrointestinal diseases, situation in Mexico. Enf Infec Microbiol. 31:137–151. 2011.

3 

World Health Organization: WHO's first ever global estimates of foodborne diseases find children under 5 account for almost one third of deaths. Geneva, Switzerland, 2015. Available from: https://www.who.int.

4 

World Health Organization: WHO publishes list of bacteria for which new antibiotics are urgently needed. Geneva, Switzerland, 2017. Available from: https://www.who.int.

5 

Reis RS and Horn F: Enteropathogenic Escherichia coli, Samonella, Shigella and Yersinia: Cellular aspects of host-bacteria interactions in enteric diseases. Gut Pathog. 2(8)2010.PubMed/NCBI View Article : Google Scholar

6 

Zaheer R, Cook SR, Barbieri R, Goji N, Cameron A, Petkau A, Polo RO, Tymensen L, Stamm C, Song J, et al: Surveillance of Enterococcus spp reveals distinct species and antimicrobial resistance diversity across a one-health continuum. Sci Rep. 10(3937)2020.PubMed/NCBI View Article : Google Scholar

7 

Fariñas MC and Martínez-Martínez L: Multiresistant gram-negative bacterial infections: Enterobacteria, pseudomonas aeruginosa, Acinetobacter baumannii and other non-fermenting gram-negative bacilli. Enferm Infecc Microbiol Clin. 31:402–409. 2013.PubMed/NCBI View Article : Google Scholar : (In Spanish).

8 

Silva-Díaz H, Bustamante-Canelo O, Aguilar-Gamboa F, Mera-Villasis K, Ipanaque-Chozo J, Seclen-Bernabe E and Vergara-Espinosa M: Predominant enteropathogens in acute diarrhea and associated variables in children at the lambayeque regional hospital, Peru. Horiz Med. 17:38–44. 2017.

9 

Oliva-Menacho J, Oliva-Candela J and Garcia-Hjarles M: Multi-drug resistant bacteria isolated from medical stethoscopes in a level III hospital. Rev Med Hered. 28:242–246. 2017.

10 

Hernandez del Sol CR and Vazquez Hernandez G, Mesa Delgado Z, Bermudez Aleman RI, Sotolongo Rodriguez Y and Vazquez Hernandez G: Enteropathogenic bacteria associated with acute diarrheal disease in children. Acta Médica del Centro. 11:28–34. 2017.

11 

López-Pueyo MJ, Barcenilla-Gaite F, Amaya-Villar R and Garnacho-Montero J: Antibiotic multiresistance in critical care units. Med Intensiva. 35:41–53. 2011.PubMed/NCBI View Article : Google Scholar : (In Spanish).

12 

Torres C, Alonso CA, Ruiz-Ripa L, León-Sampedro R, del Campo R and Coque TM: Antimicrobial Resistance in Enterococcus spp. of animal origin. In: Antimicrobial Resistance in Bacteria from Livestock and Companion Animals. Schwarz S, Cavaco LM and Shen J (eds). Wiley Online Library, USA, pp185-227, 2018.

13 

Levin-Reisman I, Brauner A, Ronin I and Balaban NQ: Epistasis between antibiotic tolerance, persistence, and resistance mutations. Proc Natl Acad Sci USa. 116:14734–14739. 2019.PubMed/NCBI View Article : Google Scholar

14 

Fernández-García L, Fernandez-Cuenca F, Blasco L, López-Rojas R, Ambroa A, Lopez M, Pascual Á, Bou G and Tomás M: Relationship between tolerance and persistence mechanisms in Acinetobacter baumannii strains with AbkAB toxin-antitoxin system. Antimicrob Agents Chemother. 62:e00250–18. 2018.PubMed/NCBI View Article : Google Scholar

15 

Pacios O, Blasco L, Bleriot I, Fernandez-Garcia L, González Bardanca M, Ambroa A, López M, Bou G and Tomás M: Strategies to combat multidrug-resistant and persistent infectious diseases. Antibiotics (Basel). 9(65)2020.PubMed/NCBI View Article : Google Scholar

16 

Brauner A, Fridman O, Gefen O and Balaban NQ: . Distinguishing between resistance, tolerance and persistence to antibiotic treatment. Nat Rev Microbiol. 14:320–330. 2016.PubMed/NCBI View Article : Google Scholar

17 

Remes Troche JM: Reflections on antibiotic resistance and what to do about it. Rev Gastroenterol Mex. 81:1–2. 2016.PubMed/NCBI View Article : Google Scholar : (In English, Spanish).

18 

Thapa Shrestha U, Adhikari N, Maharjan R, Banjara MR, Rijal KR, Basnyat SR and Agrawal VP: Mutildrug resistant Vibrio cholerae O1 from clinical and environmental samples in Kathmandu city. BMC Infect Dis. 15(104)2015.PubMed/NCBI View Article : Google Scholar

19 

Hawkey PM, Warren RE, Livermore DM, McNulty CAM, Enoch DA, Otter JA and Wilson A: Treatment of infections caused by multidrug-resistant gram-negative bacteria: Report of the British society for antimicrobial chemotherapy/healthcare infection society/British infection association joint working party. J Antimicrob Chemother. 73 (Suppl 3):iii2–iii78. 2018.PubMed/NCBI View Article : Google Scholar

20 

Alkofide H, Alhammad AM, Alruwaili A, Aldemerdash A, Almangour TA, Alsuwayegh A, Almoqbel D, Albati A, Alsaud A and Enani M: Multidrug-resistant and extensively drug-resistant Enterobacteriaceae: Prevalence, treatments, and outcomes-a retrospective cohort study. Infect Drug Resist. 13:4653–4662. 2020.PubMed/NCBI View Article : Google Scholar

21 

Nadeem SF, Gohar UF, Tahir SF, Mukhtar H, Pornpukdeewattana S, Nukthamna P, Moula Ali AM, Bavisetty S and Masa S: Antimicrobial resistance: More than 70 years of war between humans and bacteria. Crit Rev Microbiol. 46:578–599. 2020.PubMed/NCBI View Article : Google Scholar

22 

Wiedenbeck J and Cohan FM: Origins of bacterial diversity through horizontal genetic transfer and adaptation to new ecological niches. FEMS Microbiol Rev. 35:957–976. 2011.PubMed/NCBI View Article : Google Scholar

23 

Toft C and Andersson SGE: Evolutionary microbial genomics: Insights into bacterial host adaptation. Nat Rev Genet. 11:465–475. 2010.PubMed/NCBI View Article : Google Scholar

24 

Bliven KA and Maurelli AT: Evolution of bacterial pathogens within the human host. Microbiol Spectr 4: 10.1128/microbiolspec.VMBF-0017-2015, 2016.

25 

Soler N and Forterre P: Vesiduction: The fourth way of HGT. Environ Microbiol. 22:2457–2460. 2020.PubMed/NCBI View Article : Google Scholar

26 

Griffith F: The significance of pneumococcal types. J Hyg (Lond). 27:113–159. 1928.PubMed/NCBI View Article : Google Scholar

27 

Lederberg J and Tatum EL: Gene recombination in Escherichia coli. Nature. 158(558)1946.PubMed/NCBI View Article : Google Scholar

28 

Zinder ND and Lederberg J: Genetic exchange in Salmonella. J Bacteriol. 64:679–699. 1952.PubMed/NCBI View Article : Google Scholar

29 

Domingues S, Nielsen KM and da Silva GJ: Various pathways leading to the acquisition of antibiotic resistance by natural transformation. Mob Genet Elements. 2:257–260. 2012.PubMed/NCBI View Article : Google Scholar

30 

Calderón G and Aguilar L: Antimicrobial resistance: More resistant microorganisms and antibiotics. Rev Méd Costa Rica Centroam. 73:757–763. 2016.

31 

Ellison CK, Dalia TN, Vidal Ceballos A, Wang JC, Biais N, Brun YV and Dalia AB: Retraction of DNA-bound type IV competence pili initiates DNA uptake during natural transformation in Vibrio cholerae. Nat Microbiol. 3:773–780. 2018.PubMed/NCBI View Article : Google Scholar

32 

Graf FE, Palm M, Warringer J and Farewell A: Inhibiting conjugation as a tool in the fight against antibiotic resistance. Drug Dev Res. 80:19–23. 2019.PubMed/NCBI View Article : Google Scholar

33 

Krenek P, Samajova O, Luptovciak I, Doskocilova A, Komis G and Samaj J: Transient plant transformation mediated by Agrobacterium tumefaciens: Principles, methods and applications. Biotechnol Adv. 33:1024–1042. 2015.PubMed/NCBI View Article : Google Scholar

34 

Bitto NJ, Chapman R, Pidot S, Costin A, Lo C, Choi J, D'Cruze T, Reynolds EC, Dashper SG, Turnbull L, et al: Bacterial membrane vesicles transport their DNA cargo into host cells. Sci Rep. 7(7072)2017.PubMed/NCBI View Article : Google Scholar

35 

Parkinson JS: Classic spotlight: The discovery of bacterial transduction. J Bacteriol. 198:2899–2900. 2016.PubMed/NCBI View Article : Google Scholar

36 

Abebe E, Tegegne B and Tibebu S: A review on molecular mechanisms of bacterial resistance to antibiotics. Eur J Appl Sci. 8:301–310. 2016.

37 

Balcázar JL: Implications of bacteriophages on the acquisition and spread of antibiotic resistance in the environment. Int Microbiol. 23:475–479. 2020.PubMed/NCBI View Article : Google Scholar

38 

Brown L, Wolf JM, Prados-Rosales R and Casadevall A: Through the wall: Extracellular vesicles in gram-positive bacteria, mycobacteria and fungi. Nat Rev Microbiol. 13:620–630. 2015.PubMed/NCBI View Article : Google Scholar

39 

Schwechheimer C and Kuehn MJ: Outer-membrane vesicles from gram-negative bacteria: Biogenesis and functions. Nat Rev Microbiol. 13:605–619. 2015.PubMed/NCBI View Article : Google Scholar

40 

Acosta RG and Vargas CM: Bacterial resistance mechanism. Diagnóstico. 57:82–86. 2018.

41 

Reygaert WC: An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 4:482–501. 2018.PubMed/NCBI View Article : Google Scholar

42 

Cox G and Wright GD: Intrinsic antibiotic resistance: Mechanisms, origins, challenges and solutions. Int J Med Microbiol. 303:287–292. 2013.PubMed/NCBI View Article : Google Scholar

43 

Martinez JL: General principles of antibiotic resistance in bacteria. Drug Discov Today Technol. 11:33–39. 2014.PubMed/NCBI View Article : Google Scholar

44 

Bébéar CM and Pereyre S: Mechanisms of drug resistance in Mycoplasma pneumoniae. Curr Drug Targets Infect Disord. 5:263–271. 2005.PubMed/NCBI View Article : Google Scholar

45 

Zhao F, Liu J, Shi W, Huang F, Liu L, Zhao S and Zhang J: Antimicrobial susceptibility and genotyping of Mycoplasma pneumoniae isolates in Beijing, China, from 2014 to 2016. Antimicrob Resist Infect Control. 8(18)2019.PubMed/NCBI View Article : Google Scholar

46 

O'Shea R and Moser HE: Physicochemical properties of antibacterial compounds: Implications for drug discovery. J Med Chem. 51:2871–2878. 2008.PubMed/NCBI View Article : Google Scholar

47 

Davies J and Davies D: Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev. 74:417–433. 2010.PubMed/NCBI View Article : Google Scholar

48 

Martinez JL and Baquero F: Mutation frequencies and antibiotic resistance. Antimicrob Agents Chemother. 44:1771–1777. 2000.PubMed/NCBI View Article : Google Scholar

49 

Binet R and Maurelli AT: Frequency of spontaneous mutations that confer antibiotic resistance in Chlamydia spp. Antimicrob Agents Chemother. 49:2865–2873. 2005.PubMed/NCBI View Article : Google Scholar

50 

Wang G, Wilson TJM, Jiang Q and Taylor DE: Spontaneous mutations that confer antibiotic resistance in Helicobacter pylori. Antimicrob Agents Chemother. 45:727–733. 2001.PubMed/NCBI View Article : Google Scholar

51 

Coculescu BI: Antimicrobial resistance induced by genetic changes. J Med Life. 2:114–123. 2009.PubMed/NCBI

52 

Meštrović T, Virok DP, Ljubin-Sternak S, Raffai T, Burián K and Vraneš J: Antimicrobial resistance screening in Chlamydia trachomatis by optimized McCoy cell culture system and direct qPCR-based monitoring of chlamydial growth. Methods Mol Biol. 2042:33–43. 2019.PubMed/NCBI View Article : Google Scholar

53 

Kondrashov FA: Gene duplication as a mechanism of genomic adaptation to a changing environment. Proc Biol Sci. 279:5048–5057. 2012.PubMed/NCBI View Article : Google Scholar

54 

Calçada C, Silva M, Baptista V, Thathy V, Silva-Pedrosa R, Granja D, Ferreira PE, Gil JP, Fidock DA and Veiga MI: Expansion of a specific Plasmodium falciparum PfMDR1 haplotype in southeast Asia with increased substrate transport. mBio. 11:e02093–20. 2020.PubMed/NCBI View Article : Google Scholar

55 

Munita JM and Arias CA: Mechanisms of antibiotic resistance. Microbiol Spectr 4: 10.1128/microbiolspec. VMBF-0016-2015, 2016.

56 

Begum S, Begum T, Rahman N and Khan RA: A review on antibiotic resistance and way of combating antimicrobial resistance. GSC Biol Pharm Sci. 14:87–97. 2021.

57 

Etebu E and Arikekpar I: Antibiotics: Classification and mechanisms of action with emphasis on molecular perspectives. Int J Appl Microbiol Biotechnol Res. 4:90–101. 2016.

58 

Poulikakos P, Tansarli GS and Falagas ME: Combination antibiotic treatment versus monotherapy for multidrug-resistant, extensively drug-resistant, and pandrug-resistant Acinetobacter infections: A systematic review. Eur J Clin Microbiol Infect Dis. 33:1675–1685. 2014.PubMed/NCBI View Article : Google Scholar

59 

Kapoor G, Saigal S and Elongavan A: Action and resistance mechanisms of antibiotics: A guide for clinicians. J Anaesthesiol Clin Pharmacol. 33:300–305. 2017.PubMed/NCBI View Article : Google Scholar

60 

Epand RM, Walker C, Epand RF and Magarvey NA: Molecular mechanisms of membrane targeting antibiotics. Biochim Biophys Acta. 1858:980–987. 2016.PubMed/NCBI View Article : Google Scholar

61 

Hooper DC and Jacoby GA: Topoisomerase inhibitors: Fluoroquinolone mechanisms of action and resistance. Cold Spring Harb Perspect Med. 6(a025320)2016.PubMed/NCBI View Article : Google Scholar

62 

Santajit S and Indrawattana N: Mechanisms of antimicrobial resistance in ESKAPE pathogens. Biomed Res Int. 2016(2475067)2016.PubMed/NCBI View Article : Google Scholar

63 

Troncoso C, Pavez M, Santos A, Salazar R and Barrientos Díaz L: Structural and physiological implications of bacterial cell in antibiotic resistance mechanisms. Int J Morphol. 35:1214–1223. 2017.

64 

Zhou G, Shi QS, Huang XM and Xie XB: The three bacterial lines of defense against antimicrobial agents. Int J Mol Sci. 16:21711–21733. 2015.PubMed/NCBI View Article : Google Scholar

65 

Borges A, Abreu AC, Dias C, Saavedra MJ, Borges F and Simões M: New perspectives on the use of phytochemicals as an emergent strategy to control bacterial infections including biofilms. Molecules. 21(877)2016.PubMed/NCBI View Article : Google Scholar

66 

Pérez-Cano H and Robles-Contreras A: Basic aspects of the mechanisms of bacterial resistance. Rev Med MD. 4:186–191. 2013.

67 

Sager M, Benten WP, Engelhardt E, Gougoula C and Benga L: Characterization of biofilm formation in [Pasteurella] pneumotropica and [Actinobacillus] muris isolates of mouse origin. PLoS One. 10(e0138778)2015.PubMed/NCBI View Article : Google Scholar

68 

Martinez JL, Vicente MF, Delgado-Iribarren A, Perez-Diaz JC and Baquero F: Small plasmids are involved in amoxicillin-clavulanate resistance in Escherichia coli. Antimicrob Agents Chemother. 33(595)1989.PubMed/NCBI View Article : Google Scholar

69 

Sideraki V, Huang W, Palzkill T and Gilbert HF: A secondary drug resistance mutation of TEM-1 beta-lactamase that suppresses misfolding and aggregation. Proc Natl Acad Sci USA. 98:283–288. 2001.PubMed/NCBI View Article : Google Scholar

70 

Salverda ML, De Visser JA and Barlow M: Natural evolution of TEM-1 β-lactamase: experimental reconstruction and clinical relevance. FEMS Microbiol Rev. 34:1015–1036. 2010.PubMed/NCBI View Article : Google Scholar

71 

Delgado-Valverde M, Sojo-Dorado J, Pascual A and Rodríguez-Baño J: Clinical management of infections caused by multidrug-resistant Enterobacteriaceae. Ther Adv Infect Dis. 1:49–69. 2013.PubMed/NCBI View Article : Google Scholar

72 

Rodríguez-Baño J, Gutiérrez-Gutiérrez B, Machuca I and Pascual A: Treatment of infections caused by extended-spectrum-beta-lactamase-, AmpC-, and carbapenemase-producing Enterobacteriaceae. Clin Microbiol Rev. 31:e00079–17. 2018.PubMed/NCBI View Article : Google Scholar

73 

Retamar P, Portillo MM, López-Prieto MD, Rodríguez-López F, de Cueto M, García MV, Gómez MJ, Del Arco A, Muñoz A, Sánchez-Porto A, et al: Impact of inadequate empirical therapy on the mortality of patients with bloodstream infections: A propensity score-based analysis. Antimicrob Agents Chemother. 56:472–478. 2012.PubMed/NCBI View Article : Google Scholar

74 

Nørgaard SM, Jensen CS, Aalestrup J, Vandenbroucke-Grauls CM, de Boer MG and Pedersen AB: Choice of therapeutic interventions and outcomes for the treatment of infections caused by multidrug-resistant gram-negative pathogens: A systematic review. Antimicrob Resist Infect Control. 8(170)2019.PubMed/NCBI View Article : Google Scholar

75 

Falagas ME, Karageorgopoulos DE and Nordmann P: Therapeutic options for infections with Enterobacteriaceae producing carbapenem-hydrolyzing enzymes. Future Microbiol. 6:653–666. 2011.PubMed/NCBI View Article : Google Scholar

76 

Gill EE, Franco OL and Hancock REW: Antibiotic adjuvants: Diverse strategies for controlling drug-resistant pathogens. Chem Biol Drug Des. 85:56–78. 2015.PubMed/NCBI View Article : Google Scholar

77 

Lin DM, Koskella B and Lin HC: Phage therapy: An alternative to antibiotics in the age of multi-drug resistance. World J Gastrointest Pharmacol Ther. 8:162–173. 2017.PubMed/NCBI View Article : Google Scholar

78 

Reina J and Reina N: Phage theraphy, an alternative to antibiotic therapy? Rev Esp Quimioter. 31:101–104. 2018.PubMed/NCBI(In Spanish).

79 

Castañeda GCD: Intestinal microbota, probiotics and prebiotics. Enferm Inv (Ambato). 2:156–160. 2017.

80 

Játiva-Mariño E, Manterola C, Macias R and Narváez D: Probiotics and Prebiotics: Its role in childhood acute diarrheal disease therapy. Int J Morphol. 39:294–301. 2021.

81 

Olveira G and González-Molero I: An update on probiotics, prebiotics and symbiotics in clinical nutrition. Endocrinol Nutr. 63:482–494. 2016.PubMed/NCBI View Article : Google Scholar : (In English, Spanish).

82 

Suárez JE: Autochthonous microbiota, probiotics and prebiotics. Nutr Hosp. 31 (Suppl 1):S3–S9. 2015.PubMed/NCBI View Article : Google Scholar : (In Spanish).

83 

Feria MG, Taborda NA, Hernandez JC and Rugeles MT: Effects of prebiotics and probiotics on gastrointestinal tract lymphoid tissue in hiv infected patients. Rev Med Chil. 145:219–229. 2017.PubMed/NCBI View Article : Google Scholar : (In Spanish).

84 

Pandey KR, Naik SR and Vakil BV: Probiotics, prebiotics and synbiotics-a review. J Food Sci Technol. 52:7577–7587. 2015.PubMed/NCBI View Article : Google Scholar

85 

González-Torralba A, García-Esteban C and Alós JI: Enteropathogens and antibiotics. Enferm Infecc Microbiol Clin (Engl Ed). 36:47–54. 2018.PubMed/NCBI View Article : Google Scholar : (In English, Spanish).

86 

Alcántar-Curiel MD, Blackburn D, Saldaña Z, Gayosso-Vázquez C, Iovine NM, De la Cruz MA and Girón JA: Multi-functional analysis of Klebsiella pneumoniae fimbrial types in adherence and biofilm formation. Virulence. 4:129–138. 2013.PubMed/NCBI View Article : Google Scholar

87 

Clarke SC, Haigh RD, Freestone PP and Williams PH: Virulence of enteropathogenic Escherichia coli, a global pathogen. Clin Microbiol Rev. 16:365–378. 2003.PubMed/NCBI View Article : Google Scholar

88 

Gallego-Maldonado G, Otálora-Díaz AS, Urbano-Cáceres EX and Morales-Súarez C: Bacterial multiresistance: Therapeutic challenge in renal transplantation. Univ Salud. 21:72–87. 2019.

89 

Schroeder GN and Hilbi H: Molecular pathogenesis of Shigella spp.: Controlling host cell signaling, invasion, and death by type III secretion. Clin Microbiol Rev. 21:134–156. 2008.PubMed/NCBI View Article : Google Scholar

90 

Tanwar J, Das S, Fatima Z and Hameed S: Multidrug resistance: An emerging crisis. Interdiscip Perspect Infect Dis. 2014(541340)2014.PubMed/NCBI View Article : Google Scholar

91 

Alonso-Pérez C, Alcántara-Salinas A, Escobar-Rojas V, Ramírez-Sandoval MP, Reyes-Hernández MU, Guerrero-Becerra M, Vargas-Mosso ME, Hernández-Magaña R, Anzures-Gutiérrez SA, Cuevas-López LL, et al: Gastroenteritis by Campylobacter in children. Current concepts. Bol Clin Hosp Infant Edo Son. 36:88–101. 2019.

92 

Navon-Venezia S, Kondratyeva K and Carattoli A: Klebsiella pneumoniae: A major worldwide source and shuttle for antibiotic resistance. FEMS Microbiol Rev. 41:252–275. 2017.PubMed/NCBI View Article : Google Scholar

93 

Fernández-Abreu A, Bravo-Fariñas LC, Rivero-Navea G, Nuñez-Fernández FA, Cruz-Infante Y, Águila-Sánchez A and Hernández-Martínez JL: Determination of biofilms and extended-spectrum beta-lactamases in Vibrio cholerae non-O1, non-O139 isolates from patients with diarrhea in Cuba. Rev Cubana Med Trop. 71:1–7. 2019.

94 

Riveros M and Ochoa TJ: Relevant public health enteropathogens. Rev Peru Med Exp Salud Publica. 32:157–164. 2015.PubMed/NCBI(In Spanish).

95 

Macero-Gualpa LJ, Vásquez-Véliz RM and Reyes-Sánchez RR: Wound infection by aeromona hydrophila, a case report in ecuador. Rev Med FCM-UCSG. 23:95–99. 2019.

96 

Fàbrega A and Vila J: Yersinia enterocolitica: Pathogenesis, virulence and antimicrobial resistance. Enferm Infecc Microbiol Clin. 30:24–32. 2012.PubMed/NCBI View Article : Google Scholar

97 

Liu GY: Molecular pathogenesis of Staphylococcus aureus infection. Pediatr Res. 65:71R–77R. 2009.PubMed/NCBI View Article : Google Scholar

98 

Harnisz M and Korzeniewska E: The prevalence of multidrug-resistant Aeromonas spp in the municipal wastewater system and their dissemination in the environment. Sci Total Environ. 626:377–383. 2018.PubMed/NCBI View Article : Google Scholar

99 

Fiore E, Van Tyne D and Gilmore MS: Pathogenicity of enterococci. Microbiol Spectr. 7:10.1128/microbiolspec.GPP3-0053-2018. 2019.

Related Articles

Journal Cover

December-2022
Volume 24 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
González-Villarreal JA, González-Lozano KJ, Aréchiga-Carvajal ET, Morlett-Chávez JA, Luévanos-Escareño MP, Balagurusamy N and Salinas-Santander MA: Molecular mechanisms of multidrug resistance in clinically relevant enteropathogenic bacteria (Review). Exp Ther Med 24: 753, 2022
APA
González-Villarreal, J.A., González-Lozano, K.J., Aréchiga-Carvajal, E.T., Morlett-Chávez, J.A., Luévanos-Escareño, M.P., Balagurusamy, N., & Salinas-Santander, M.A. (2022). Molecular mechanisms of multidrug resistance in clinically relevant enteropathogenic bacteria (Review). Experimental and Therapeutic Medicine, 24, 753. https://doi.org/10.3892/etm.2022.11689
MLA
González-Villarreal, J. A., González-Lozano, K. J., Aréchiga-Carvajal, E. T., Morlett-Chávez, J. A., Luévanos-Escareño, M. P., Balagurusamy, N., Salinas-Santander, M. A."Molecular mechanisms of multidrug resistance in clinically relevant enteropathogenic bacteria (Review)". Experimental and Therapeutic Medicine 24.6 (2022): 753.
Chicago
González-Villarreal, J. A., González-Lozano, K. J., Aréchiga-Carvajal, E. T., Morlett-Chávez, J. A., Luévanos-Escareño, M. P., Balagurusamy, N., Salinas-Santander, M. A."Molecular mechanisms of multidrug resistance in clinically relevant enteropathogenic bacteria (Review)". Experimental and Therapeutic Medicine 24, no. 6 (2022): 753. https://doi.org/10.3892/etm.2022.11689