ArticlePDF Available

Mechanisms of Antibiotics Resistance in Bacteria

Authors:

Abstract

The resistance is determining a growing hug issue in health fields worldwide, where the bacterial cells possessed the ability to resist the old antibiotics as well as the newly discovered antibiotics through several capabilities and mechanisms, including the natural, acquired and cross ones, this paper will highlight most of the antibiotics and the mechanics of resistance.
Sys Rev Pharm 2020; 11(6): 817 823
A multifaceted review journal in the field of pharmacy
E-ISSN 0976-2779 P-ISSN 0975-8453
817 Systematic Review Pharmacy Vol 11, Issue 6, 2020
Mechanisms of Antibiotics Resistance in Bacteria
Thualfakar Hayder Hasan1*, Raad A. Al-Harmoosh1,2
1Department of Medical Laboratories Techniques; Altoosi University College, Najaf, Iraq.
2College of Science; University of Kufa, Najaf, Iraq.
E-mail: thualfakar@altoosi.edu.iq, raad.alharmoosh@altoosi.edu.iq
Article History: Submitted: 13.04.2020 Revised: 17.05.2020 Accepted: 24.06.2020
ABSTRACT
The resistance is determining a growing hug issue in health fields
worldwide, where the bacterial cells possessed the ability to resist the
old antibiotics as well as the newly discovered antibiotics through
several capabilities and mechanisms, including the natural, acquired
and cross ones, this paper will highlight most of the antibiotics and the
mechanics of resistance.
Keywords: Resistance, Antibiotics, Bacteria, QRDRs, Efflux pumps
Correspondence:
Thualfakar Hayder Hasan
Department of Medical Laboratories Techniques, Altoosi University
College, Najaf, Iraq
E-mail: thulafakar@altoosi.edu.iq
DOI: 10.31838/srp.2020.6.118
@Advanced Scientific Research. All rights reserved
INTRODUCTION
Bacterial resistance is the capability of bacterial cells to
prevent antibiotic bacteriostatic or bactericidal effects [1].
The excessive and unintended usage of antibiotics
contributes to resistance development in bacteria [2].
Because of the extensive uptake, the evolvement
of microorganisms resistant with the time and problems
have arisen with these resistant microorganisms for the
treatment of certain infections [3]. Nowadays, resistance is
determining as a big issue in the path of new drug synthesis,
developing antibiotic resistance is a major public health
problem worldwide [4].
The four principal forms of antibiotic resistance evolve as
1- Natural resistance (Intrinsic, Structural)
In this type of resistance, the usage of antibiotics is not
associated with the resistance but it caused by the bacteria's
structural properties [5]. This occurs as a result of intrinsic
resistance, or microorganism which doesn't follow the target
antibiotic structure, or antibiotics which due to its
characteristics do not encounter its target [6]. Gram-
negative bacteria and vancomycin, for example, vancomycin
antibiotics does not move through the outer membrane soo
that these Gram-negative bacteria are naturally insusceptible
to vancomycino[7].
Likewise, L-form bacteria that are cell wall-less types of the
bacteria, such a Ureaplasma and Mycoplasma Mycoplasma
that are naturally owning beta-lactam antibiotics resistance
[8].
2. Acquired resistance
Regardless of resistance development due to alteration in
the genetic features of bacteria, an acquired because it is not
affected by the antibiotics it was previously susceptible to it
[9]. This form of resistance comes from the main
chromosome or extra chromosome structures (plasmids,
transposons, etc.) [10].
Chromosomal resistance results from mutations that change
randomly bacterial chromosome, these mutations can occur
by certain physical and chemical factors [11].
This may be due to changes in the composition of bacterial
cells, so that may be decreased bacterial drug permeability,
or maybe changes to the drug's target in the cell [12].
Streptomycin, aminoglycosides,o erythromycin,o, and
lincomycin can develop resistance to these forms [13].
Extrachromosomal resistance relies on extrachromosomal
genetic materials that can be transmitted via plasmids,
transposons, and integrons [14]. Plasmidsoareosegments of
DNAothatocanoreplicateoindependentlyoof chromosomal
DNA [15]. A plasmid is typically responsible for the
development of antibiotic inactive enzymes [16].
There are main forms of holding genetic material (resistance
genes and plasmids) from bacterial cells, this form are
transduction, transformation, conjugation, and mechanism
of transposition [17].
The genes with antibiotic resistance on the chromosome or
plasmid are intertwined and are situated at the beginning
with different integration groups, or integrons.
Recombination is very normal in integrons [18].
3. Cross-resistance
It is mean the resistance to a specific antibiotic by specific
microorganisms, that work with the identical or related
mechanisms and that are also resistant to other antibiotics
[19]. This is generally seen when antibiotics have common
structures: such as resistance to erythromycin,neomycin-
kanamycin, or resistance to cephalosporins and penicillins
[20].
However, cross-resistance can some times be seen in a
completely distinct group of drugs as well, like a cross-
resistance that exists amongst erythromycin-lincomycin,
this resistance might be the chromosomal origin or not [21].
4. Multi-drug and other types of resistance
Multidrug-resistant species are typically pathogens that
have been resistant to their antibiotics, this ensures that the
bacteria will no longer be eliminated or regulated by a single
drug [22]. Inappropriate utilization of antibiotics
for treatment culminated in the introduction of multidrug-
resistant pathogenic bacteria [23]. Either of the two
mechanisms can induce multidrug resistance in bacteria
[24].
Firstly, these bacteria will acquire several genes, each coding
for specific drug resistance, this form of resistance usually
exists on R-plasmids [25].
Secondly, the form of multidrug resistance may also occur
by enhanced gene expression encoding for efflux pumps,
enzymatic inactivation for antibiotics, changes in target
structure, and others [26].
Thualfakar Hayder Hasan et al / Mechanisms of Antibiotics Resistance in Bacteria
818 Systematic Review Pharmacy Vol 11, Issue 6, 2020
If the bacterial strains are not susceptible to three or more
antimicrobial types, they are called multidrug-resistant
(MDR) bacteria. If the species, resistant to all but one or two
classes of antibiotics, are deemed highly resistant to
medicines, whether the species resistant to all usable
antibiotics are known as pan-drug resistant [27,28].
For example, Acinetobacter species with multidrug
resistance (MDR) can be identified as the bacteria that
having the resistant ability to at least three groups of
antibiotics classes, for example for all penicillin and
cephalosporin, aminoglycosides and quinolones groups
[29].
Extensive Acinetobacter spp., drug-resistant (XDR), isolate
resistant to the three types of antibiotics classes mentioned
above in (MDR), and even carbapenem-resistant,
Acinetobacter spp. , Pandrug resistant, or pan-resistant
(PDR), these bacteria can be going to be the XDR as well as
polymyxin-resistant and tigecycline resistance [30,31].
Mechanisms of Antibiotics Resistance
A-The modifications
Modifications that happen in the drug-related receptor and
the location of the target regions of the relation with the
antibiotics are distinct, these can be complex enzymes and
ribosomes [32]. The most frequently identified resistance
consistent with variations in the ribosomal target is in
macrolide antibiotics [33]. The most popular examples here
are the evolvement of penicillin resistance due to the
mutations of penicillin-binding proteins beta-lactamase
enzymes in Staphylococcus aureus, Streptococcus
pneumoniae, Neisseria meningitides, and Enterococcus
faecium strains [34].
B.i Enzymatic inactivation of antibiotics
Most of the bacteria synthesize antibiotic degrading
enzymes, the enzymatic inactivation mechanism is one of
the most important antibiotics resistance mechanisms [35].
In this group, beta-lactamases, aminoglycosidase,
chloramphenicol, and erythromycin modifying enzymes are
the most popular examples [36].
C. Reduction of the inner and outer membrane
permeability
This mechanism results from changes in the permeability of
the internal and external membrane so that decreased drug
uptake into the cell or rapidly ejected from the pump
systems [37]. Due to a decrease in membrane permeability
as a result of porin mutations that may occur in proteins of
resistant strains for example; a mutation in specific porins
called OprD can cause resistance to carbapenem in
Pseudomonas aeruginosa strain [38]. Reduction in outer
membrane permeability can play an important role in
quinolone resistance and aminoglycoside resistance [39].
D. Active Pumps System
Resistance develops most commonly in the tetracycline
group of antibiotics via the active pump systems [40]. With
an energy-dependent active pumping system, tetracyclines
are thrown out and cannot concentrate within the cell[41].
Thisomechanismooforesistanceoisoinoplasmidoandochrom
osomalocontrol.oActive pumping systems for example are
effective in resisting quinolones,o14-membered
macrolides,ochloramphenicoloandobeta-lactamso[42].
E.oUsingoanoalternativeometabolicopathway
Unlikeosomeoofotheotargetoalterationsoinobacteria, the
latest drug-susceptible pathway eliminates the need for
objective development [43]. Bacteria can prepare folic acid
from the environment, rather than synthesizing folic acid so
that it becomes resistant among sulfonamide and
trimethoprim [44].
ResistanceobyoAntibioticsogroupoMechanisms
A. Beta-lactams Resistance
Antibiotics of beta-lactam are a wide class of antibiotics,
including penicillins, cephalosporins, monobactams, and
carbapenems [45]. Synthesis of beta-lactamase enzymes is
the most common resistance mechanism here [46].
1- Beta-lactamase Enzymes
At the molecular level, there are 4 groups (A, B, C, D)of
beta-lactamase enzymeso[47]. Beta-lactamases A, C, and D
that deferent from B-class that function cool ester enzymes
mediated, while the latest was need zinc ion as
metalloenzyme [48].
Beta-lactamases Class A
These resistances occur in both Gram-positive and Gram-
negative bacteria and mostly mediated by plasmid or
transposon. Capable usually of being inducible [48]. This
group includes the gram-negative bacteria TEM, SHV,
ESBL. ESBL primarily occurs in E. coli and Klebsiella
pneumoniae [49].
Beta-lactamases Class B
Bacteroides fragilis, observable species of Aeromonas and
Legionella, enzymes that hydrolyze carbapenems, penicillin,
and cephalosporins [50].
Beta-lactamases Class C
Generally seen in Gram-negative bacteria and chromosome-
localized (Group I, AmpC, etc.) [51]. This resistance
mechanism is not inhibited by clavulanic acid and has an
inducible characteristic so produced in high levels in the
presence of beta-lactam antibiotics [52]. Often known as
Inducibleo Beta-Lactamaseso(IBL), they found in
Enterobacterocloacae,oCitrobacterofreundii,oSerratiaomarce
scens, and P. aeruginosa [53].
Beta-lactamases Class D
These enzymes are induced by beta-lactamases antibiotics
and produced in Gram-positive cocci such as Staphylococcus
aureus so that degrade Oxacillin [53].
2. Modifications in Penicillin-Binding Proteins (PBP)
Penicillin-binding proteins (PBP) in peptidoglycan
synthesis in the bac responsible for the Antibiotic target of
beta-lactam, Carboxypeptidase PBPs, and the enzymes of
Thualfakar Hayder Hasan et al / Mechanisms of Antibiotics Resistance in Bacteria
819 Systematic Review Pharmacy Vol 11, Issue 6, 2020
transpeptidase PBP is the most common in gram-positive
bacteria, due to changes in it, resistance results in [54].
Methicillin-resistant S. aureus (MRSA) is willing to take
responsibility for methicillin resistance in strains, mecA
gene, this gene results in PBP-2a synthesis enhancing beta-
lactam antibiotic resistance [55]. The modifications in S.
pneumoniae in PBP 2b are responsible for the resistance to
penicillin and cephalosporin [56].
3. Modifications in Proteins of the membrane
Change in the porin channels in gram-negative bacteria,o
for example, P. aeruginosa with a devoted channel protein
registered in OprD may evolve carbapenem resistance [57].
Antibiotic accumulation can be prevented in the active
pump systems cell. Consequently, the group of beta-lactams,
tetracyclines, chloramphenicol, and quinolones can lead to
resistance [58].
B. Antibiotics Resistance of Aminoglycoside Group
1. Aminoglycosides Modifying Enzymes
The most important mechanism for the emergence of
resistance to aminoglycosides in aerobic gram-negative
bacteria is enzymatic inactivation. Enzyme modifying has a
major role in resistance to aminoglycosides [59]. These
enzymes are often of plasmid or transposon origin, there are
acetyltransferase and phosphotransferase in this group [60].
Modified enzymes are responsible for the high extent of
gentamicin resistanceoinoenterococcio[61].
2. Ribosomal target Modifications
This approach is crucial in Streptomycin resistance, the
target of streptomycin is not connected to the ribosomal 30S
subunit due to mutations in the ribosomal 30S, in
enterococci, this kind of resistance to streptomycin is
essential [62].
C. The resistance of Tetracyclines
1. Prevention of the absorption of drugs into cells and
active pump systems
Reduction of membrane permeability resulting from
spontaneous chromosome mutations in bacteria as a result
of resistance development to prevent drug uptake [63]. The
organisms also can develop Tetracyclines resistance
depending on active pump systems [64].
2. Protection of Ribosome
The second significant mechanism which leads to
tetracycline resistance [65]. With tetM, tetO, tetQ, tetS genes
inhibit drug activity by modifying a cytoplasmic ribosome
that binds to the tetracycline [66]. These genes have been
found in many genera like Campylobacter, Mycoplasma,
Ureaplasma, and Bacteroides, for example. They are plasmid
and chromosome origin [67].
D. The resistance of Macrolide, lincosamide,
streptogramins (MLS)groups
Gram-negative bacteria are naturally resistant to MLS
group antibiotics
1. Ribosomal Target Modification
This mechanism is most common in Gram-Positive
bacteria, in the 50S ribosomal subunit, this is connected to
the drug with the 23S of the ribosome in rRNA-specific
methylation of an adenine molecule has a structural change
and reduces the drug's binding to ribosomal RNA. The
resistance is of a structural or inducible type [68,69].
2. Inactivation of drug by Enzymatic activity
The bacterial cells having enzymes that play a critical role in
resistance like Erythromycin and other Macrolides
resistance [70].
E. The resistance of Chloramphenicol
The inactivation of the chloramphenicol acetyltransferase
(CAT) by enzymes that acetylate the chloramphenicol
antibiotic leads to resistance in bacteria produced by this
enzyme [71]. Reduced drug uptake in certain bacteria
especially gram-negative can also be responsible for
chloramphenicol resistance [72].
F. The resistance of Quinolones
There are different mechanisms for quinolone resistance
that including
1. Mutation modification of the target topoisomerase
Modifications in the target enzymes topoisomerases
caused mainly by mutations that reduce the affinity of
quinolones without compromising the enzyme function are
the most common mechanism of acquired quinolone
resistance and have already been reported in several
bacterial species [73,74]. resistance-related mutations are
clustered in discrete regions of the enzyme subunits, called
regions determining quinolone resistance (QRDRs) [75].
2. A decreased intake of drugs by reduced permeability or
active efflux
Increased resistance to quinolones in gram-negative bacteria
due to variations in their outer membrane proteins so that
they reduce the intake of drugs [76].
3. The target protection of topoisomerase with specific
proteins
Target protection is provided by a family of small
pentapeptide-repeat proteins, called Qnr proteins, which
bind to the targets for topoisomerase and protect them from
quinolone interaction [77]. A similar mechanism has
developed in bacteria to protect topoisomerases from
microcin, which are pentapeptide-repeat family proteins
that are produced as a mechanism of biological competition
by certain bacteria and can kill susceptible bacteria by
inhibiting their topoisomerases [78].
4. Inactivation of the drug
The most recently identified mechanism of resistance to
quinolones was inactivation by drug modification [79].
Acetylation is performed by a plasmid-encoded AAC
enzyme variant which, has the ability to acetylate some
quinolone molecules in addition to aminoglycosides and
have unsubstituted secondary amines such as ciprofloxacin
and norfloxacin [80].
Thualfakar Hayder Hasan et al / Mechanisms of Antibiotics Resistance in Bacteria
820 Systematic Review Pharmacy Vol 11, Issue 6, 2020
G. Resistance of Rifampicin
The high-level resistance develops readily mainly due to
chromosomal mutation in most bacteria so developed of
stable changes that prevent binding in RNA polymerase
[81]. Rifampicin should only be used in association with
another antibacterial drug since the mutation risk is high.
Rifampicin resistance is not transferable, and other
antibacterials do not have cross-resistance [82,83].
H. Resistance of Sulfonamide and Trimethoprim
Sulfonamides are para-aminobenzoic acid analogs (PABA)
and the dihydropteroate synthesis (DHPS)enzyme and
trimethoprim dihydrofolate reductase (DHFR) metabolic
pathways inhibiting tetrahydrofolic acid synthesis in
bacteria [84,85]. Chromosomal and plasmid-mediated
resistance to sulfonamides and trimethoprim [86].
Bacterialoexpressionoof the DHPSosulfonamides low-
affinity plasmid comprising this case is the most commonly
observed resistance to sulfonamide [87,88].
FUNDING SUPPORT
This research did not receive any specific grant from
funding agencies in the public, commercial, or not-for-
profit sectors.
CONFLICT OF INTEREST
None to declare.
ETHICAL CLEARANCE
All data was approved and carried out in accordance with
approved guidelines.
REFERENCES
1. Munita, J. M., and Arias, C. A. 2016. Mechanisms of
Antibiotic Resistance. Microbiology spectrum, 4(2),
10.1128/microbiolspec.VMBF-0016-2015.
https://doi.org/10.1128/microbiolspec.VMBF-0016-
2015
2. Kraemer, S. A., Ramachandran, A., and Perron, G. G.
2019. Antibiotic Pollution in the Environment: From
Microbial Ecology to Public
Policy. Microorganisms, 7(6),180.
https://doi.org/10.3390/microorganisms7060180
3. Hayder, T., and Aljanaby, A. A. J. 2019a.Antibiotics
susceptibility patterns of Citrobacter freundii isolated
from pa-tients with urinary tract infection in Al-Najaf
governorate Iraq. International Journal of Research
in Pharmaceutical Sciences, 10(2), 1481 1488.
https://doi.org/10.26452/ijrps.v10i2.722
4. Hayder, T., and Aljanaby, A. A. J. 2019b. Genotypic
Characterization of Antimicrobial Resistance-
Associated Genes in Citrobacter Freundii Isolated
from Patients with Urinary Tract Infection in Al-Najaf
Governorate-Iraq. OnLine Journal of Biological
Sciences, 19(2), 132 145.
https://doi.org/10.3844/ojbsci.2019.132.145
5. Kadhum, H. A., and Hasan, T. H. 2019. The Study of
Bacillus Subtils Antimicrobial Activity on Some of the
Pathological Isolates. International Journal of Drug
Delivery Technology, 9(02), 193-196.
6. Waglechner, N., and Wright, G. D. 2017. Antibiotic
. BMC
biology, 15(1), 1-8.
7. Antonoplis, A., Zang, X., Wegner, T., Wender, P. A.,
and Cegelski, L. 2019. Vancomycin arginine
conjugate inhibits growth of carbapenem-resistant E.
coli and targets cell-wall synthesis. ACS chemical
biology, 14(9), 2065-2070.
8. Claessen, D., and Errington, J. 2019. Cell Wall
Deficiency as a Coping Strategy for Stress. Trends in
microbiology.
9. Andersson, D. I., Balaban, N. Q., Baquero, F.,
Courvalin, P., Glaser, P., Gophna, U., ... and Tønjum,
T. 2020. Antibiotic resistance: turning evolutionary
principles into clinical reality. FEMS Microbiology
Reviews.
10. Aljanaby, A. A. J., and Aljanaby, I. A. J. 2018.
Prevalence of aerobic pathogenic bacteria isolated
from patients with burn infection and their
antimicrobial susceptibility patterns in Al-Najaf City,
Iraq-a three-year cross-sectional
study. F1000Research, 7(1157), 1157.
11. Majeed, H. T., and Aljanaby, A. A. J. 2019. Antibiotic
Susceptibility Patterns and Prevalence of Some
Extended Spectrum Beta-Lactamases Genes in Gram-
Negative Bacteria Isolated from Patients Infected with
Urinary Tract Infections in Al-Najaf City,
Iraq. Avicenna journal of medical biotechnology, 11(2),
192.
12. AL-Harmoosh, R. A. , Jarallah, E. M. , AL-Shamari,
A.M and AL-khafaji, H. M. 2017.Detection of Efflux
Pumps Genes in Clinical Isolates of Acinetobacter
baumannii. Research J. Pharm. and Tech.
10(12):4231-4236.
13. Krause, K. M., Serio, A. W., Kane, T. R., and
Connolly, L. E. 2016. Aminoglycosides: An
Overview. Cold Spring Harbor perspectives in
medicine, 6(6), a027029.
https://doi.org/10.1101/cshperspect.a027029
14. Jabuk, S. I.A. , jabuk, N.A.G. , AL-Harmoosh, R. A.
and Hussien, R.S.2017.Isolation and Identification of
antibiotic producing bacteria from local soil in
Babylon province. Euphrates Journal of Agriculture
Science / Second Veterinary Conference, 304-3010.
15. Thomas, C. M., and Frost, L. S. 2014. Plasmid
Genomes, Introduction to. Molecular Life Sciences, 1
20. https://doi.org/10.1007/978-1-4614-6436-5_107-2.
16. Aljanaby A.A., Witwit I.N., Mubark H.M., Al-Labban
H.M.Y.2019. synthesis and characterization of new
imidazole azo ligand with some of transition metal
ions, and their biological effect on two pathogenic
bacteria of burn patients. International Journal of
Research in Pharmaceutical Sciences 10 (3), 1847-
1856, 2019.
17. Aljanaby, A.A.J.,Adam, R.W.,Al-Labban H.M.Y.,
Kadhim I.N.2019. Synthesis, characterization, and
antibacterial activity of some new pyrimidine
Thualfakar Hayder Hasan et al / Mechanisms of Antibiotics Resistance in Bacteria
821 Systematic Review Pharmacy Vol 11, Issue 6, 2020
derivatives from chalcone derivatives. Drug Invention
Today 11 (7), 1732-1739.
18. Adam R.W., Al-Labban H.M.Y., Aljanaby A.A.J.,
Abbas N.A.2019. Synthesis, characterization and
antibacterial activity of some new of novel 1,2,3-
triazole-chalcone derivatives from N-acetyl-
5Hdibenzo [B,F] azepine-5-carboxamide. Nano
Biomed. Eng 11 (2), 99-110.
19. Etebu, E., and Arikekpar, I. 2016. Antibiotics:
Classification and mechanisms of action with
emphasis on molecular perspectives. Int. J. Appl.
Microbiol. Biotechnol. Res, 4(2016), 90-101.
20. Jahne, M. A., Rogers, S. W., Ramler, I. P., Holder, E.,
and Hayes, G. 2015. Hierarchal clustering yields
insight into multidrug-resistant bacteria isolated from
a cattle feedlot wastewater treatment
system. Environmental monitoring and
assessment, 187(1), 4168.
21. Ng, H. F. 2019. Selection And Characterization Of A
Tigecycline-Resistant Mutant Of Mycobacterium
Abscessus To Identify Possible Resistance
Determinants (Doctoral dissertation, UTAR).
22. Alanis, A. J. 2005. Resistance to antibiotics: are we in
the post-antibiotic era?. Archives of medical
research, 36(6), 697-705.
23. Dheda, K., Gumbo, T., Maartens, G., Dooley, K. E.,
McNerney, R., Murray, M., ... and Theron, G. 2017.
The epidemiology, pathogenesis, transmission,
diagnosis, and management of multidrug-resistant,
extensively drug-resistant, and incurable
tuberculosis. The lancet Respiratory medicine, 5(4),
291-360.
24. Rello, J., Bunsow, E., and Perez, A. 2016. What if
there were no new antibiotics? A look at
alternatives. Expert review of clinical
pharmacology, 9(12), 1547-1555.
25. Tóth, S., Szepesi, Á., Tran-Nguyen, V. K., Sarkadi, B.,
Német, K., Falson, P., ... and Boumendjel, A. 2020.
Synthesis and Anticancer Cytotoxicity of Azaaurones
Overcoming Multidrug Resistance. Molecules, 25(3),
764.
26. Salloum, S., Michel, T. A. W. K., and Tayyara, L.
2020. Bacterial resistance to antibiotics and associated
factors in two hospital centers in Lebanon from
January 2017 to June 2017. Infection Prevention in
Practice, 2(2), 100043.
27. Stanford, K., Zaheer, R., Klima, C., McAllister, T.,
Peters, D., Niu, Y. D., and Ralston, B. 2020.
Antimicrobial Resistance in Members of the Bacterial
Bovine Respiratory Disease Complex Isolated from
Lung Tissue of Cattle Mortalities Managed with or
without the Use of Antimicrobials. Microorganisms,
8(2), 288.
28. AL-Harmoosh , R.A. and Jarallah, E.M. 2015.First
Detection of The blaNDM-1 and blaNDM-2 Genes in
a Clinical Isolates of Acinetobacter baumannii in
Hillah Hospitals-Iraq. International Journal of
Advanced Research . 3(10) : 1407 -1416.
29. Escudero, J. A., Loot, C., and Mazel, D. 2018.
Integrons as adaptive devices. In Molecular
Mechanisms of Microbial Evolution (pp. 199-239).
Springer, Cham.
30. AL-Harmoosh , R.A. , Jarallah, E.M. and AL-Shamari,
A.M. 2016. Coexistence of the blaIMP and blaSIM
Genes in Clinical Isolates of Acinetobacter baumannii
In Babylon Hospitals-Iraq. International Journal of
PharmTech Research.9(7) : 257-264.
31. Nikaido, H. 2009. Multidrug resistance in
bacteria. Annual review of biochemistry, 78, 119-146.
32. Prashanth, K., Vasanth, T., Saranathan, R., Makki, A.
R., and Pagal, S. 2012. Antibiotic resistance, biofilms
and quorum sensing in Acinetobacter
species. Antibiotic resistant bacteria: a continuous
challenge in the new millennium, 179-212.
33. Shaikh, S. A., Jain, T., Sandhu, G., Latha, N., and
Jayaram, B. 2007. From drug target to leads-sketching
a physicochemical pathway for lead molecule design
in silico. Current pharmaceutical design, 13(34), 3454-
3470.
34. Southon, S. B., Beres, S. B., Kachroo, P., Saavedra, M.
O., Erlendsdottir, H., Haraldsson, G., ... and
Kristinsson, K. G. 2020. Population genomic
molecular epidemiological study of macrolide
resistant Streptococcus pyogenes in Iceland, 1995-
2016: Identification of a large clonal population with a
pbp2x mutation conferring reduced in vitro beta-
lactam susceptibility. bioRxiv.
35. Pérez-Llarena, F. J., and Bou, G. 2016. Proteomics as
a tool for studying bacterial virulence and
antimicrobial resistance. Frontiers in microbiology, 7,
410.
36. Sharkey, L. K., and O'Neill, A. J. 2019. Molecular
Mechanisms of Antibiotic Resistance Part
II. Bacterial Resistance to Antibiotics From Molecules
to Man, 27-50.
37. Santajit, S., and Indrawattana, N. 2016. Mechanisms
of antimicrobial resistance in ESKAPE
pathogens. BioMed research international, 2016.
38. Nikaido, H., and Pagès, J. M. 2012. Broad-specificity
efflux pumps and their role in multidrug resistance of
Gram-negative bacteria. FEMS microbiology
reviews, 36(2), 340 363.
https://doi.org/10.1111/j.1574-6976.2011.00290.x
39. Li, H., Luo, Y. F., Williams, B. J., Blackwell, T. S., and
Xie, C. M. 2012. Structure and function of OprD
protein in Pseudomonas aeruginosa: from antibiotic
resistance to novel therapies. International Journal of
Medical Microbiology, 302(2), 63-68.
40. Breidenstein, E. B., de la Fuente-Núñez, C., and
Hancock, R. E. 2011. Pseudomonas aeruginosa: all
roads lead to resistance. Trends in microbiology, 19(8),
419-426.
41. Li, T., Liu, C., Lu, J., Gaurav, G. K., and Chen, W.
2020. Determination of how tetracycline influences
nitrogen removal performance, community structure,
and functional genes of biofilm systems. Journal of the
Taiwan Institute of Chemical Engineers, 106, 99-109.
Thualfakar Hayder Hasan et al / Mechanisms of Antibiotics Resistance in Bacteria
822 Systematic Review Pharmacy Vol 11, Issue 6, 2020
42. Guo, R., Li, K., Qin, J., Niu, S., and Hong, W. 2020.
Development of Polycationic Micelles as an Efficient
Delivery System of Antibiotics Overcoming Biological
Barriers to Reverse Multidrug Resistance in
Escherichia coli. Nanoscale.
43. Fatahi-Bafghi, M. 2019. Antibiotic resistance genes in
the Actinobacteria phylum. European Journal of
Clinical Microbiology and Infectious Diseases, 1-26.
44. Tan, J., Tay, J., Hedrick, J., and Yang, Y. Y. 2020.
Synthetic macromolecules as therapeutics that
overcome resistance in cancer and microbial
infection. Biomaterials, 120078.
45. Fernández-Villa, D., Aguilar, M. R., and Rojo, L.
2019. Folic Acid Antagonists: Antimicrobial and
Immunomodulating Mechanisms and
Applications. International journal of molecular
sciences, 20(20), 4996.
46. Zango, U. U., Ibrahim, M., and Shawai, S. A. A. 2019.
-lactam antibiotic drug resistance. MOJ
Drug Des Develop Ther, 3(2), 52-58.
47. Heinz, E., Ejaz, H., Scott, J. B., Wang, N., Gujaran, S.,
Pickard, D., ... and Strugnell, R. A. 2019. Resistance
mechanisms and population structure of highly drug
resistant Klebsiella in Pakistan during the
introduction of the carbapenemase NDM-1. Scientific
reports, 9(1), 1-13.
48. Walkty, A., Karlowsky, J. A., Baxter, M. R., Adam, H.
J., Alexander, D., Bay, D. C., ... and Zhanel, G. G.
2020. Fosfomycin resistance mediated by fos genes
remains rare among extended-spectrum beta-
lactamase-producing Escherichia coli clinical isolates
recovered from the urine of patients evaluated at
Canadian hospitals (CANWARD, 2007
2017). Diagnostic Microbiology and Infectious
Disease, 96(3), 114962.
49. Lomovskaya, O., Nelson, K., Rubio-Aparicio, D.,
Tsivkovski, R., Sun, D., and Dudley, M. N. 2020.
Impact of Intrinsic Resistance Mechanisms on
Potency of QPX7728, a New Ultrabroad-Spectrum
Beta-Lactamase Inhibitor of Serine and Metallo-Beta-
Lactamases in Enterobacteriaceae, Pseudomonas
aeruginosa, and Acinetobacter
baumannii. Antimicrobial Agents and
Chemotherapy, 64(6).
50. De Oliveira, D. M., Forde, B. M., Kidd, T. J., Harris, P.
N., Schembri, M. A., Beatson, S. A., ... and Walker,
M. J. 2020. Antimicrobial Resistance in ESKAPE
Pathogens. Clinical Microbiology Reviews, 33(3).
51. Jacobs, M. R., Abdelhamed, A. M., Good, C. E.,
Rhoads, D. D., Hujer, K. M., Hujer, A. M., ... and
Kreiswirth, B. N. 2019. ARGONAUT-I: activity of
cefiderocol (S-649266), a siderophore cephalosporin,
against gram-negative bacteria, including
carbapenem-resistant nonfermenters and
Enterobacteriaceae with defined extended-spectrum
-lactamases and carbapenemases. Antimicrobial
agents and chemotherapy, 63(1), e01801-18.
52. Meini, M. R., Llarrull, L. I., and Vila, A. J. 2015.
Overcoming differences: the catalytic mechanism of
metallo- -lactamases. FEBS letters, 589(22), 3419-
3432.
53. Philippon, A., Arlet, G., and Jacoby, G. A. 2002.
Plasmid-determined AmpC-type beta-
lactamases. Antimicrobial agents and
chemotherapy, 46(1), 1 11.
https://doi.org/10.1128/aac.46.1.1-11.2002
54. Kong, K. F., Schneper, L., and Mathee, K. 2010. Beta-
lactam antibiotics: from antibiosis to resistance and
bacteriology. APMIS : acta pathologica,
microbiologica, et immunologica Scandinavica, 118(1),
1 36. https://doi.org/10.1111/j.1600-
0463.2009.02563.x
55. Akata, K., Muratani, T., Yatera, K., Naito, K.,
Noguchi,
Induction of plasmid- -lactamase
DHA- -
lactams in Enterobacteriaceae. PLOS ONE, 14(7),
e0218589.
https://doi.org/10.1371/journal.pone.0218589
56. Fisher, J. F., and -Lactam
Resistance Mechanisms: Gram-Positive Bacteria
andMycobacterium tuberculosis. Cold Spring Harbor
Perspectives in Medicine, 6(5), a025221.
https://doi.org/10.1101/cshperspect.a025221
57. Sauvage, E., and Terrak, M. 2016.
Glycosyltransferases and Transpeptidases/Penicillin-
Binding Proteins: Valuable Targets for New
Antibacterials. Antibiotics, 5(1), 12.
https://doi.org/10.3390/antibiotics5010012
58. Harkins, C. P., Pichon, B., Doumith, M., Parkhill, J.,
Westh, H., Tomasz, A., de Lencastre, H., Bentley, S.
D., Kearns, A. M., and Holden, M. 2017. Methicillin-
resistant Staphylococcus aureus emerged long before
the introduction of methicillin into clinical
practice. Genome biology, 18(1), 130.
https://doi.org/10.1186/s13059-017-1252-9
59. Naha, A., Miryala, S. K., Debroy, R., Ramaiah, S., and
Anbarasu, A. 2020. Elucidating the multi-drug
resistance mechanism of Enterococcus faecalis V583:
A gene interaction network analysis. Gene, 144704.
60. Gonzalez, M. R., Ducret, V., Leoni, S., and Perron, K.
2019. Pseudomonas aeruginosa zinc homeostasis: Key
issues for an opportunistic pathogen. Biochimica et
Biophysica Acta (BBA)-Gene Regulatory
Mechanisms, 1862(7), 722-733.
61. Gil-Gil, T., Martínez, J. L., and Blanco, P. 2020.
Mechanisms of antimicrobial resistance in
Stenotrophomonas maltophilia: a review of current
knowledge. Expert Review of Anti-infective
Therapy, 18(4), 335-347.
62. Breijyeh, Z., Jubeh, B., and Karaman, R. 2020.
Resistance of Gram-Negative Bacteria to Current
Antibacterial Agents and Approaches to Resolve
It. Molecules, 25(6), 1340.
63. Das, B., Verma, J., Kumar, P., Ghosh, A., and
Ramamurthy, T. 2020. Antibiotic resistance in Vibrio
cholerae: understanding the ecology of resistance
genes and mechanisms. Vaccine, 38, A83-A92.
Thualfakar Hayder Hasan et al / Mechanisms of Antibiotics Resistance in Bacteria
823 Systematic Review Pharmacy Vol 11, Issue 6, 2020
64. Böhm, M. E., Razavi, M., Marathe, N. P., Flach, C. F.,
and Larsson, D. J. 2020. Discovery of a novel
integron-borne aminoglycoside resistance gene
present in clinical pathogens by screening
environmental bacterial
communities. Microbiome, 8(1), 1-11.
65. Sivagami, K., Vignesh, V. J., Srinivasan, R.,
Divyapriya, G., and Nambi, I. M. 2020. Antibiotic
usage, residues and resistance genes from food
animals to human and environment: An Indian
scenario. Journal of Environmental Chemical
Engineering, 8(1), 102221.
66. Kapoor, G., Saigal, S., and Elongavan, A. 2017.
Action and resistance mechanisms of antibiotics: A
guide for clinicians. Journal of anaesthesiology, clinical
pharmacology, 33(3), 300.
67. Poole, K. 2007. Efflux pumps as antimicrobial
resistance mechanisms. Annals of medicine, 39(3),
162-176.
68. Linkevicius, M., Sandegren, L., and Andersson, D. I.
2016. Potential of tetracycline resistance proteins to
evolve tigecycline resistance. Antimicrobial agents and
chemotherapy, 60(2), 789-796.
69. Zhanel, G. G., Esquivel, J., Zelenitsky, S., Lawrence, C.
K., Adam, H. J., Golden, A., ... and Bay, D. 2020.
Omadacycline: A Novel Oral and Intravenous
Aminomethylcycline Antibiotic Agent. Drugs, 1-29.
70. Cesur, S., and Demiröz, A. P. 2013. Antibiotics and
the mechanisms of resistance to antibiotics. Medical
Journal of Islamic World Academy of
Sciences, 109(1007), 1-5.
71. Siibak, T. 2011. Effect of antibiotics on ribosome
assembly is indirect (Doctoral dissertation).
72. Petrov, A., Meskauskas, A., and Dinman, J. D. 2004.
Ribosomal protein L3: influence on ribosome
structure and function. RNA biology, 1(1), 58-64.
73. Munita, J. M., and Arias, C. A. 2016. Mechanisms of
antibiotic resistance. Virulence mechanisms of
bacterial pathogens, 481-511.
74. Roberts, M. C., and Schwarz, S. 2017. Tetracycline
and chloramphenicol resistance mechanisms.
In Antimicrobial drug resistance (pp. 231-243).
Springer, Cham.
75. Santajit, S., and Indrawattana, N. 2016. Mechanisms
of antimicrobial resistance in ESKAPE
pathogens. BioMed research international, 2016.
76. Theuretzbacher, U., Bush, K., Harbarth, S., Paul, M.,
Rex, J. H., Tacconelli, E., and Thwaites, G. E. 2020.
Critical analysis of antibacterial agents in clinical
development. Nature Reviews Microbiology, 1-13.
77. Bansal, S., Bajaj, P., Pandey, S., and Tandon, V. 2017.
Topoisomerases: Resistance versus sensitivity, how far
we can go?. Medicinal research reviews, 37(2), 404-438.
78. Makarewicz, O., Klinger-Strobel, M., Ehricht, R.,
Kresken, M., and Pletz, M. W. 2017. Antibiotic
resistance in pulmonary infections: mechanisms and
epidemiology. Anti-infectives and the Lung, 21.
79. Ghai, I., and Ghai, S. 2017. Exploring bacterial outer
membrane barrier to combat bad bugs. Infection and
drug resistance, 10, 261.
80. Ruiz, J., Pons, M. J., and Gomes, C. 2012.
Transferable mechanisms of quinolone
resistance. International journal of antimicrobial
agents, 40(3), 196-203.
81. Hooper, D. C., and Jacoby, G. A. 2016.
Topoisomerase inhibitors: fluoroquinolone
mechanisms of action and resistance. Cold Spring
Harbor perspectives in medicine, 6(9), a025320.
82. El-Badawy, M. F., Tawakol, W. M., El-Far, S. W.,
Maghrabi, I. A., Al-Ghamdi, S. A., Mansy, M. S., ...
and Shohayeb, M. M. 2017. Molecular identification
of aminoglycoside-modifying enzymes and plasmid-
mediated quinolone resistance genes among Klebsiella
pneumoniae clinical isolates recovered from Egyptian
patients. International journal of microbiology, 2017.
83. Sithole, B. 2015. Peptide-antibiotic conjugates as novel
developments for increased antimicrobial transport and
efficiency (Doctoral dissertation, University of York).
84. Goldstein, B. P. 2014. Resistance to rifampicin: a
review. The Journal of antibiotics, 67(9), 625-630.
85. Imran, M., Das, K. R., and Naik, M. M. 2019. Co-
selection of multi-antibiotic resistance in bacterial
pathogens in metal and microplastic contaminated
environments: An emerging health
threat. Chemosphere, 215, 846-857.
86. Capasso, C., and Supuran, C. T. 2019.
Dihydropteroate synthase (sulfonamides) and
dihydrofolate reductase inhibitors. Bacterial Resistance
to Antibiotics From Molecules to Man, 163-172.
87. and Adley, C. 2017.
Antibiotics: mode of action and mechanisms of
resistance. Formatex Research Center: Badajoz, Spain,
536-545.
88. Ricken, B., Kolvenbach, B. A., Bergesch, C., Benndorf,
D., Kroll, K., Strnad, H., ... and Schäffer, A. 2017.
FMNH 2-dependent monooxygenases initiate
catabolism of sulfonamides in Microbacterium sp.
strain BR1 subsisting on sulfonamide
antibiotics. Scientific reports, 7(1), 1-11.
... The two isolates (1 and 5) were isolated from the urine of the National Center for Educational Laboratories in Medical City. The third group C includes 5 bacterial isolates (14,15,20,29,34) found with genetic convergence and in one lineage and the source of isolates (14,15,20) was the urine of the National Center for Educational Laboratories in Medical City. The isolation no. ...
... The two isolates (1 and 5) were isolated from the urine of the National Center for Educational Laboratories in Medical City. The third group C includes 5 bacterial isolates (14,15,20,29,34) found with genetic convergence and in one lineage and the source of isolates (14,15,20) was the urine of the National Center for Educational Laboratories in Medical City. The isolation no. ...
Article
Fifty isolates of Staphylococcus aureus bacteria were obtained out of 200 samples collected from clinical sources. The highest resistance to S. aureus bacteria was 100% Benzylpericillin, Oxacillin and Moxiflaxocin antibiotics, 86% Levoflaxacin resistance, 76% Erythromycin resistance, 66% Tetracycline resistance, 64% Tobramycin resistance, 62% Gentamicin resistance, 60% Clindamycin resistance and 58% Trimethoprim/Sulfamethoxazole resistance. It was 38% sensitive to Gentamicin and Clindamycin, 36% sensitive to Tobramycin, 22% sensitive to Erythromycin and 14% sensitive to Levoflaxacin while the results showed the presence of five different sized types of DNA packets when detecting Coa gene of S.aureus bacteria using PCR techniques.
... The development of defense mechanisms by bacteria against drugs that were once efficient at treating infections is known as antibiotic resistance (Blair et al., 2015). Bacterial defense armamentarium against antibiotics contributing to antibiotic resistance has evolved over time and includes the following methods: 1) genetic mutations; 2) modification of the drug target; 3) porin mutations causing a reduction in permeability; 4) increase in the number of efflux pumps; 5) enhance the secretion of inactivating enzymes and/or hydrolases; 6) changes in cell morphology; 7)Metabolic regulation or auxotrophy; 8) initiation of self-repair systems within the bacteria; 9) interaction between resistance protein and antibiotic target; 10) acquisition of resistance genes from other bacteria termed as community cooperative resistance; 11) biofilm formation and protection; 12) antibiotic avoidance (Hasan & Al-Harmoosh, 2020;Uddin et al., 2021;F. Zhang & Cheng, 2022). ...
Article
Full-text available
The rise of multi-drug resistant (MDR) pathogens poses a significant challenge to the field of infectious disease treatment. To overcome this problem, novel strategies are being explored to enhance the effectiveness of antibiotics. Antibiotic adjuvants have emerged as a promising approach to combat MDR pathogens by acting synergistically with antibiotics.
... One of the main concerns of modern infectious disease is the growing incidence of antibiotic resistance in bacteria (WHO 2017;Larsson and Flach 2022). The excessive and inappropriate use of antibiotics in humans and animals has led to the development of different defense strategies in many bacterial species, resulting in higher resistance to standard therapies being less effective and even ineffective (Hansan et al. 2020). Furthermore, in recent decades, the inappropriate use of antibiotics to control disease in livestock has increased bacterial resistance (Zhao et al. 2021). ...
Article
Full-text available
Fliou J, Spinola F, Riffi O, Zriouel A, Amechrouq A, Nalbone L, Giuffrida A, Giarratana F. 2023. Phytochemical evaluation and in-vitro antibacterial activity of ethanolic extracts of Moroccan Lavandula x intermedia leaves and flowers. Biodiversitas 24: 5788-5796. This study performed a preliminary evaluation of the phytochemical composition and in vitro antibacterial activity of ethanolic extracts of Lavandula x intermedia leaves and flowers collected in the Fez-Meknes region of Morocco. Phytochemical analyses comprised qualitative colorimetric determinations of alkaloids, anthraquinones, and terpenes and quantitative analysis of total polyphenols, flavonoids, and condensed tannins by UV spectrophotometer. Antibacterial activity was evaluated by determining minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values against different ATCC bacterial strains. The phytochemical analysis showed a high amount of total polyphenols, flavonoids, and tannins in the leaf extract and a higher amount of terpenes based on colorimetric reaction than the flower extract. A positive colorimetric reaction for alkaloids and anthraquinones was detected for both extracts. The antibacterial activity of leaves and flower extract was not different against Gram-positive and Gram-negative strains (p<0.05). The results of the present study suggest the possible use of ethanolic extracts of L. x intermedia collected in the Fez-Meknes region of Morocco as a natural agent against bacterial pathogens.
... [3][4][5] Unfortunately, indiscriminate antibiotic use, improper prescription practices, their excessive deployment in agriculture, a dearth of novel antibiotic discoveries, and the formidable challenges associated with regulatory approval have collectively led to the emergence and proliferation of antibiotic-resistant bacterial strains. [6][7][8] This proliferation of antibiotic-resistant bacteria stems from improper and excessive antibiotic usage, unsuitable prescription practices, the widespread application of antibiotics in agriculture, a lack of innovative solutions, and regulatory hurdles. In 2017, the World Health Organization (WHO) identified a group of critical disease-causing microbes, collectively known as ESKAPE bacteria, including Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp. 1 Urgent research is imperative to discover novel treatments for these bacteria, as they have developed the ability to evade various therapies and transfer their resistance to other organisms. ...
Article
Full-text available
Introduction: The primary aim of the current investigation is to assess the chemical composition and antibacterial efficacy of Ptychotis verticillata essential oil (PVEO) against three Gram-positive bacteria (Staphylococcus aureus, Micrococcus luteus, and Bacillus subtilis) and three Gram-negative bacteria (Escherichia coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae). Furthermore, this research endeavors to investigate the potential synergistic effects of PVEO when combined with established antibiotics (Amoxicillin, Erythromycin, and Ampicillin), with a focus on discerning their interaction dynamics. Methods: The phytochemical profiling of PVEO was performed via the Gas Chromatography-Mass Spectrometry technique. The broth dilution test determined the interaction effect between PVEO and these three antibiotics. Results: Components γ-terpinene (38.96%), p-cymene (19.38%), thymol (18.07%), and carvacrol (6.99%) are the major bioactive moleculs composing this studied essential oil. PVEO demonstrated significant antibacterial activity with minimum inhibitory concentration (MIC) values ranging from 0.2 against E coli to 2 mg/mL against M luteus. The assessment of the synergistic activity between PVEO and antibiotics was accomplished through the utilization of the Fractional Inhibitory Concentration Index (FICI). The combination of PVEO and amoxicillin against M luteus demonstrated the best synergistic action with a FICI value of 0.43. Conclusion: Significant reductions, ranging from two to sevenfold, in the MIC values of PVEO, and antibiotics were observed. This noteworthy synergistic interaction between highly potent essential oils and synthetic antibiotics holds the potential to open avenues for novel combination therapies aimed at addressing infections caused by multiresistant microorganisms, even at notably reduced concentrations within the pharmaceutical sector.
... Throughout the last few decades, resistance to a wide spectrum of antibiotics has been acquired by strains originating from "classical" E. coli in the previous few decades, which is a cause for concern 9 . As a result of increased antibiotic resistance, common illnesses like urinary tract infections (UTIs) have grown resistant to treatment, while more serious diseases like pneumonia and bacteremia have become progressively life-threatening 10 . The purpose of this study was to evaluate certain antibiotic resistance in E. coli isolated from pregnant women with UTIs in Najaf, Iraq. ...
Article
Full-text available
BACKGROUND: Antibiotic resistance mediated by efflux pumps is a serious public health threat. Esomeprazole magnesium trihydrate (EMT) is a proton pump inhibitor with reported antimicrobial activity, but its effects against efflux-mediated resistance are unknown. OBJECTIVE: To evaluate the antimicrobial activity of EMT alone and in combination with the efflux pump inhibitor reserpine against clinical isolates of _Pseudomonas aeruginosa, Acinetobacter baumannii_ and _Staphylococcus aureus_. METHODS: Minimum inhibitory concentration (MIC) assays were performed using the broth microdilution method on 15 non-duplicate clinical isolates and reference strains with/without EMT and reserpine. RESULTS: EMT demonstrated activity against all isolates, with MIC ranges of 4-32 μg/mL, 8-64 μg/mL and 2-16 μg/mL respectively. EMT MICs decreased 4-8-fold for 13/15 isolates when combined with reserpine, indicating EMT may inhibit efflux pumps. Similar reductions occurred for comparator antibiotics. CONCLUSIONS: This study provides the first evidence that EMT possesses intrinsic antimicrobial activity against these pathogens and may function as both an efflux pump substrate and inhibitor. EMT warrants further investigation as a potential adjuvant antibiotic for overcoming efflux-mediated resistance.
Article
Full-text available
Acinetobacter baumannii is a Gram-negative bacterium characterized by its short, round, rod-shaped morphology. It is an opportunistic pathogen that poses a significant threat, particularly to immunocompromised patients, often those with hospital stays lasting less than 90 days. Between June 2022 and July 2023, 214 urine samples were collected from individuals suspected of having urinary tract infections (UTIs). These samples were subjected to antibiotic resistance testing, focusing on detecting specific genes related to carbapenem resistance, namely blaNDM, blaKPC, and blaVIM.The study's results revealed a notable trend in antibiotic resistance among the bacterial isolates. Ceftazidime, cefotaxime, and ceftriaxone, commonly used antibiotics for UTIs, showed a high resistance rate among the tested isolates. This resistance highlights the challenges healthcare professionals face when treating UTIs caused by Acinetobacter baumannii. On the other hand, the isolates displayed a comparatively lower resistance rate to imipenem and meropenem, two necessary carbapenem antibiotics. This lower resistance to carbapenems is encouraging as these drugs are often considered the last line of defense against multidrug-resistant bacterial infections. The presence of carbapenem resistance genes, such as blaNDM, blaKPC, and blaVIM, in the Acinetobacter baumannii isolates is of particular concern. These genes confer resistance to carbapenem antibiotics, crucial for treating severe infections caused by multidrug-resistant bacteria. In conclusion, the study aims to study the growth of antibiotic resistance in Acinetobacter baumannii, especially in urinary tract infections in immunocompromised patients with more extended hospital stays. It also highlights the need for Surveys and periodic examinations to detect the spread of bacteria and their resistance. Keywords: Carbapenems, UTI, genes, blaNDM, blaKPC, and blaVIM.
Article
Full-text available
The discovery of microRNAs (miRNAs) has revolutionized our understanding of gene regulation and its impact on cellular processes. miRNAs are small non-coding RNA molecules that play a crucial role in post-transcriptional gene regulation. They are involved in various physiological and pathological processes, including development, differentiation, and disease progression. Understanding the biogenesis, mechanisms of action, and circulation of miRNAs is essential for unraveling their functional significance and potential applications in diagnostics and therapeutics. This article provides a comprehensive overview of microRNA biogenesis, mechanisms of action, and their circulation, highlighting their role in cellular regulation and their diagnostic and therapeutic potential.
Article
Full-text available
Antimicrobial resistance represents an enormous global health crisis and one of the most serious threats humans face today. Some bacterial strains have acquired resistance to nearly all antibiotics. Therefore, new antibacterial agents are crucially needed to overcome resistant bacteria. In 2017, the World Health Organization (WHO) has published a list of antibiotic-resistant priority pathogens, pathogens which present a great threat to humans and to which new antibiotics are urgently needed the list is categorized according to the urgency of need for new antibiotics as critical, high, and medium priority, in order to guide and promote research and development of new antibiotics. The majority of the WHO list is Gram-negative bacterial pathogens. Due to their distinctive structure, Gram-negative bacteria are more resistant than Gram-positive bacteria, and cause significant morbidity and mortality worldwide. Several strategies have been reported to fight and control resistant Gram-negative bacteria, like the development of antimicrobial auxiliary agents, structural modification of existing antibiotics, and research into and the study of chemical structures with new mechanisms of action and novel targets that resistant bacteria are sensitive to. Research efforts have been made to meet the urgent need for new treatments; some have succeeded to yield activity against resistant Gram-negative bacteria by deactivating the mechanism of resistance, like the action of the β-lactamase Inhibitor antibiotic adjuvants. Another promising trend was by referring to nature to develop naturally derived agents with antibacterial activity on novel targets, agents such as bacteriophages, DCAP(2-((3-(3,6-dichloro-9H-carbazol-9-yl)-2-hydroxypropyl)amino)-2(hydroxymethyl)propane1,3-diol, Odilorhabdins (ODLs), peptidic benzimidazoles, quorum sensing (QS) inhibitors, and metal-based antibacterial agents.
Article
Full-text available
Background: New antibiotic resistance determinants are generally discovered too late, long after they have irreversibly emerged in pathogens and spread widely. Early discovery of resistance genes, before or soon after their transfer to pathogens could allow more effective measures to monitor and reduce spread, and facilitate genetics-based diagnostics. Results: We modified a functional metagenomics approach followed by in silico filtering of known resistance genes to discover novel, mobilized resistance genes in class 1 integrons in wastewater-impacted environments. We identified an integron-borne gene cassette encoding a protein that conveys high-level resistance against aminoglycosides with a garosamine moiety when expressed in E. coli. The gene is named gar (garosamine-specific aminoglycoside resistance) after its specificity. It contains none of the functional domains of known aminoglycoside modifying enzymes but bears characteristics of a kinase. By searching public databases, we found that the gene is present in two sequenced clinical multi-resistant isolates (Pseudomonas aeruginosa and Luteimonas sp.) from Italy and China, respectively, but it has escaped discovery until now. Conclusion: To the best of our knowledge, this is the first time a resistance gene present in pathogens isolated from patients has been discovered by exploring the environmental microbiome. The gar gene has spread horizontally to different species in at least two continents, further limiting treatment options for bacterial infections. Its specificity to garosamine-containing aminoglycosides may reduce the usefulness of the newest semisynthetic aminoglycoside plazomicin, which is designed to avoid common aminoglycoside resistance mechanisms. Since the gene appears to be not yet common in the clinics, the data presented here enables early surveillance and maybe even mitigation of its spread.
Article
Full-text available
: Bacterial, protozoan and other microbial infections share an accelerated metabolic rate. In order to ensure a proper functioning of cell replication and proteins and nucleic acids synthesis processes, folate metabolism rate is also increased in these cases. For this reason, folic acid antagonists have been used since their discovery to treat different kinds of microbial infections, taking advantage of this metabolic difference when compared with human cells. However, resistances to these compounds have emerged since then and only combined therapies are currently used in clinic. In addition, some of these compounds have been found to have an immunomodulatory behavior that allows clinicians using them as anti-inflammatory or immunosuppressive drugs. Therefore, the aim of this review is to provide an updated state-of-the-art on the use of antifolates as antibacterial and immunomodulating agents in the clinical setting, as well as to present their action mechanisms and currently investigated biomedical applications.
Article
Synthetic macromolecular antimicrobials have shown efficacy in the treatment of multidrug resistant (MDR) pathogens. These synthetic macromolecules, inspired by Nature's antimicrobial peptides (AMPs), mitigate resistance by disrupting microbial cell membrane or targeting multiple intracellular proteins or genes. Unlike AMPs, these polymers are less prone to degradation by proteases and are easier to synthesize on a large scale. Recently, various studies have revealed that cancer cell membrane, like that of microbes, is negatively charged, and AMPs can be used as anticancer agents. Nevertheless, efforts in developing polymers as anticancer agents has remained limited. This review highlights the recent advancement in the development of synthetic biodegradable antimicrobial polymers (e.g. polycarbonates, polyesters and polypeptides) and anticancer macromolecules including peptides and polymers. Additionally, strategies to improve their in vivo bioavailability and selectivity towards bacteria and cancer cells are examined. Lastly, future perspectives, including use of artificial intelligence or machine learning, in the development of antimicrobial and anticancer macromolecules are discussed.
Article
Resistance to antibiotics have created havoc around the globe due to the emergence of multi-drug resistant (MDR) pathogenic bacterial strains. To decipher this problem, a detailed understanding of the antimicrobial resistance (AMR) genes and their resistant mechanisms are obligatory. The present study is mainly focused on an opportunistic, nosocomial bacterial strain Enterococcus faecalis V583, which possess acquired exogenous AMR genes portraying resistance against Chloramphenicol, Tetracycline, Vancomycin, Linezolid, Ampicillin and other antibiotics. An interaction network of eight AMR genes along with 40 functional partners have been constructed and analysed. Functional enrichment analysis highlighted 20, 21 and 22 genes having significant roles in Cellular Component (CC), Molecular Functions (MF) and Biological Process (BP) respectively. Clustering analysis resulted in four densely interconnected clusters (C1-C4) which were associated with three AMR mechanisms that include the alteration in drug target (pbps, mur and van genes), complete replacement/bypass of target sites (van genes) and ATP Binding Cassette (ABC) transporter efflux pump mechanisms (msrA, EF_1680, EF_1682 and pbps). Our results showed that the genes responsible for β-lactams resistance (pbp1A, 1C, 2A, 2B); glycopeptide resistance (ddl, vanBHBRBSBWXYB); erythromycin, macrolides, lincosamide and streptogramin-B (MLSB) resistance (msrA, EF_1680, EF_1682) along with mur genes (murABBCDEFG) played an important role in MDR mechanisms. Network analysis has shown the genes mraY, pbpC, murE, murG and murD possessed 26, 24, 23, 22 and 22 interactions respectively. With more number of direct interactions, these genes can be considered as hub genes that could be exploited as potential drug targets for new drug discovery.
Article
The antibacterial agents currently in clinical development are predominantly derivatives of well-established antibiotic classes and were selected to address the class-specific resistance mechanisms and determinants that were known at the time of their discovery. Many of these agents aim to target the antibiotic-resistant priority pathogens listed by the WHO, including Gram-negative bacteria in the critical priority category, such as carbapenem-resistant Acinetobacter, Pseudomonas and Enterobacterales. Although some current compounds in the pipeline have exhibited increased susceptibility rates in surveillance studies that depend on geography, pre-existing cross-resistance both within and across antibacterial classes limits the activity of many of the new agents against the most extensively drug-resistant (XDR) and pan-drug-resistant (PDR) Gram-negative pathogens. In particular, cross-resistance to unrelated classes may occur by co-selection of resistant strains, thus leading to the rapid emergence and subsequent spread of resistance. There is a continued need for innovation and new-class antibacterial agents in order to provide effective therapeutic options against infections specifically caused by XDR and PDR Gram-negative bacteria. New antibacterial agents are urgently needed to address the global increase in resistance. In this Review, Theuretzbacher and colleagues critically review the current published literature and publicly available information on antibacterial agents in all phases of clinical development.
Chapter
The discovery, commercialization, and routine administration of antimicrobial compounds to treat infections revolutionized modern medicine and changed the therapeutic paradigm. Indeed, antibiotics have become one of the most important medical interventions needed for the development of complex medical approaches such as cutting-edge surgical procedures, solid organ transplantation, and management of patients with cancer, among others. Unfortunately, the marked increase in antimicrobial resistance among common bacterial pathogens is now threatening this therapeutic accomplishment, jeopardizing the successful outcomes of critically ill patients. In fact, the World Health Organization has named antibiotic resistance as one of the three most important public health threats of the 21st century (1).
Article
Introduction: Stenotrophomonas maltophilia is a prototype of bacteria intrinsically resistant to antibiotics. The reduced susceptibility of this microorganism to antimicrobials mainly relies on the presence in its chromosome of genes encoding efflux pumps and antibiotic inactivating enzymes. Consequently, the therapeutic options for treating S. maltophilia infections are limited. Areas covered: Known mechanisms of intrinsic, acquired and phenotypic resistance to antibiotics of S. maltophilia and the consequences of such resistance for treating S. maltophilia infections are discussed. Acquisition of some genes, mainly those involved in co-trimoxazole resistance, contributes to acquired resistance. Mutation, mainly in the regulators of chromosomally-encoded antibiotic resistance genes, is a major cause for S. maltophilia acquisition of resistance. The expression of some of these genes is triggered by specific signals or stressors, which can lead to transient phenotypic resistance. Expert opinion: Treatment of S. maltophilia infections is difficult because this organism presents low susceptibility to antibiotics. Besides, it can acquire resistance to antimicrobials currently in use. Particularly problematic is the selection of mutants overexpressing efflux pumps since they present a multidrug resistance phenotype. The use of novel antimicrobials alone or in combination, together with the development of efflux pumps’ inhibitors may help in fighting S. maltophilia infections.
Article
Antibiotic resistance is one of the major challenges facing modern medicine worldwide. The past few decades have witnessed rapid progress in our understanding of the multiple factors that affect the emergence and spread of antibiotic resistance at the population level and the level of the individual patient. However, the process of translating this progress into health policy and clinical practice has been slow. Here, we attempt to consolidate current knowledge about the evolution and ecology of antibiotic resistance into a roadmap for future research as well as clinical and environmental control of antibiotic resistance. At the population level, we examine emergence, transmission and dissemination of antibiotic resistance, and at the patient level, we examine adaptation involving bacterial physiology and host resilience. Finally, we describe new approaches and technologies for improving diagnosis and treatment and minimizing the spread of resistance.
Article
Omadacycline is a novel aminomethylcycline antibiotic developed as a once-daily, intravenous and oral treatment for acute bacterial skin and skin structure infection (ABSSSI) and community-acquired bacterial pneumonia (CABP). Omadacycline, a derivative of minocycline, has a chemical structure similar to tigecycline with an alkylaminomethyl group replacing the glycylamido group at the C-9 position of the D-ring of the tetracycline core. Similar to other tetracyclines, omadacycline inhibits bacterial protein synthesis by binding to the 30S ribosomal subunit. Omadacycline possesses broad-spectrum antibacterial activity against Gram-positive and Gram-negative aerobic, anaerobic, and atypical bacteria. Omadacycline remains active against bacterial isolates possessing common tetracycline resistance mechanisms such as efflux pumps (e.g., TetK) and ribosomal protection proteins (e.g., TetM) as well as in the presence of resistance mechanisms to other antibiotic classes. The pharmacokinetics of omadacycline are best described by a linear, three-compartment model following a zero-order intravenous infusion or first-order oral administration with transit compartments to account for delayed absorption. Omadacycline has a volume of distribution (Vd) ranging from 190 to 204 L, a terminal elimination half-life (t½) of 13.5–17.1 h, total clearance (CLT) of 8.8–10.6 L/h, and protein binding of 21.3% in healthy subjects. Oral bioavailability of omadacycline is estimated to be 34.5%. A single oral dose of 300 mg (bioequivalent to 100 mg IV) of omadacycline administered to fasted subjects achieved a maximum plasma concentration (Cmax) of 0.5–0.6 mg/L and an area under the plasma concentration-time curve from 0 to infinity (AUC0–∞) of 9.6–11.9 mg h/L. The free plasma area under concentration–time curve divided by the minimum inhibitory concentration (i.e., fAUC24h/MIC), has been established as the pharmacodynamic parameter predictive of omadacycline antibacterial efficacy. Several animal models including neutropenic murine lung infection, thigh infection, and intraperitoneal challenge model have documented the in vivo antibacterial efficacy of omadacycline. A phase II clinical trial on complicated skin and skin structure infection (cSSSI) and three phase III clinical trials on ABSSSI and CABP demonstrated the safety and efficacy of omadacycline. The phase III trials, OASIS-1 (ABSSSI), OASIS-2 (ABSSSI), and OPTIC (CABP), established non-inferiority of omadacycline to linezolid (OASIS-1, OASIS-2) and moxifloxacin (OPTIC), respectively. Omadacycline is currently approved by the FDA for use in treatment of ABSSSI and CABP. Phase II clinical trials involving patients with acute cystitis and acute pyelonephritis are in progress. Mild, transient gastrointestinal events are the predominant adverse effects associated with use of omadacycline. Based on clinical trial data to date, the adverse effect profile of omadacycline is similar to studied comparators, linezolid and moxifloxacin. Unlike tigecycline and eravacycline, omadacycline has an oral formulation that allows for step-down therapy from the intravenous formulation, potentially facilitating earlier hospital discharge, outpatient therapy, and cost savings. Omadacycline has a potential role as part of an antimicrobial stewardship program in the treatment of patients with infections caused by antibiotic-resistant and multidrug-resistant Gram-positive [including methicillin-resistant Staphylococcus aureus (MRSA)] and Gram-negative pathogens.