ArticlePDF AvailableLiterature Review

Bacterial proton motive force as an unprecedented target to control antimicrobial resistance

Authors:

Abstract

Novel antibacterial therapies are urgently required to tackle the increasing number of multidrug-resistant pathogens. Identification of new antimicrobial targets is critical to avoid possible cross-resistance issues. Bacterial proton motive force (PMF), an energetic pathway located on the bacterial membrane, crucially regulates various biological possesses such as adenosine triphosphate synthesis, active transport of molecules, and rotation of bacterial flagella. Nevertheless, the potential of bacterial PMF as an antibacterial target remains largely unexplored. The PMF generally comprises electric potential (ΔΨ) and transmembrane proton gradient (ΔpH). In this review, we present an overview of bacterial PMF, including its functions and characterizations, highlighting the representative antimicrobial agents that specifically target either ΔΨ or ΔpH. At the same time, we also discuss the adjuvant potential of bacterial PMF-targeting compounds. Lastly, we highlight the value of PMF disruptors in preventing the transmission of antibiotic resistance genes. These findings suggest that bacterial PMF represents an unprecedented target, providing a comprehensive approach to controlling antimicrobial resistance.
Received: 20 April 2022
|
Revised: 10 January 2023
|
Accepted: 26 February 2023
DOI: 10.1002/med.21946
REVIEW ARTICLE
Bacterial proton motive force as an
unprecedented target to control antimicrobial
resistance
Bingqing Yang
1
|Ziwen Tong
1
|Jingru Shi
1
|
Zhiqiang Wang
1,2
|Yuan Liu
1,2,3
1
Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of
Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
2
Joint International Research Laboratory of Agriculture and AgriProduct Safety, the Ministry of Education of China, Yangzhou
University, Yangzhou, Jiangsu, China
3
Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China
Correspondence
Zhiqiang Wang and Yuan Liu, Jiangsu
Coinnovation Center for Prevention and
Control of Important Animal Infectious Diseases
and Zoonoses, College of Veterinary Medicine,
Yangzhou University, Yangzhou, Jiangsu, China.
Email: zqwang@yzu.edu.cn and
liuyuan2018@yzu.edu.cn
Funding information
National Key Research and Development
Program of China, Grant/Award Numbers:
2021YFD1801000, 2018YFA0903400; Young
Elite Scientists Sponsorship Program by CAST,
Grant/Award Number: 2020QNRC001; Priority
Academic Program Development of Jiangsu
Higher Education Institutions (PAPD),
Grant/Award Number: 2019; Jiangsu
Agricultural Science and Technology Innovation
Fund, Grant/Award Numbers: CX(20)3091,
CX(21)2010; National Natural Science
Foundation of China, Grant/Award Numbers:
32222084, 32172907, 32002331; 111 Project,
Grant/Award Number: D18007
Abstract
Novel antibacterial therapies are urgently required
to tackle the increasing number of multidrug
resistant pathogens. Identification of new antimicro-
bial targets is critical to avoid possible cross
resistance issues. Bacterial proton motive force
(PMF), an energetic pathway located on the bacterial
membrane, crucially regulates various biological pos-
sesses such as adenosine triphosphate synthesis,
active transport of molecules, and rotation of bacte-
rial flagella. Nevertheless, the potential of bacterial
PMF as an antibacterial target remains largely
unexplored. The PMF generally comprises electric
potential (ΔΨ) and transmembrane proton gradient
(ΔpH). In this review, we present an overview of
bacterial PMF, including its functions and characteri-
zations, highlighting the representative antimicrobial
agents that specifically target either ΔΨ or ΔpH. At
the same time, we also discuss the adjuvant potential
of bacterial PMFtargeting compounds. Lastly, we
highlight the value of PMF disruptors in preventing
Med Res Rev. 2023;43:10681090.wileyonlinelibrary.com/journal/med1068
|
© 2023 Wiley Periodicals LLC.
the transmission of antibiotic resistance genes. These
findings suggest that bacterial PMF represents an
unprecedented target, providing a comprehensive
approach to controlling antimicrobial resistance.
KEYWORDS
antibiotic resistance, antimicrobial agents, bacteria, proton motive
force, target
1|INTRODUCTION
Antimicrobial resistance (AMR) is an increasingly severe problem threatening global health. It is indicated that nearly
1.27 million deaths directly result from antibiotic resistance and 4.95 million deaths are closely correlated to
resistant bacterial infections in 2019.
1
It is medically urgent to develop new antimicrobial strategies with traditional
antibiotics gradually losing their effectiveness.
2
Newly discovered lead compounds hold immense potential in this
battle. However, identifying compounds targeting the repetitious intracellular physiological processes, such as
membrane and cell wall disruption, inhibition of DNA/RNA, and protein synthesis, easily forms crossresistance
with the existing drug resistance determinants, greatly hampering their clinical translation.
3
Therefore, seeking
novel targets will be conducive to expanding the current antibacterial arsenals.
4
The cell membrane, an unexplored promising target for antimicrobial agents, has gained increasing attention
because of its importance in nongrowing bacteria and the success of emerging antibiotics.
57
Several novel
membrane targets, especially membrane phospholipids, have been successfully identified recently. For example,
daptomycin displays antimicrobial activities against Staphylococcus aureus by targeting phosphatidylglycerol (PG), a
central phospholipid component, and triggering severe membrane blebbing.
8
A linear undecapeptide, SLAPS25,
potentiates multiple antibiotic efficacies through simultaneously binding to lipopolysaccharide (LPS) and PG of
drugresistant Escherichia coli membrane.
9
Additionally, two other essential phospholipids, phosphatidylethanola-
mine (PE) and cardiolipin (CL), also inspire the development of new patterns of antibiotics. Specifically, by inhibiting
CL, a microbiotaderived nonribosomal peptide termed telomycin, and its natural analogs, leads to the rapid
membrane lysis of S. aureus and Bacillus subtilis, causing a bactericidal effect.
10
Likewise, the methylation status of
the whole PE opens the premise for highlevel cinnamycin production in inducing antibacterial activities.
11
More interestingly, some unique previously unexplored elements exist on the membrane, such as menaquinone
(vitamin K2), the target of lysocin E,
12
and BamA, the target of novel antibiotic darobactin.
13
Specifically, the
binding of lysocin E with menaquinone leads to a series of membrane responses, such as potassium leakage and
membrane depolarization, thus exhibiting potent bactericidal activity against Grampositive bacteria. It is also
noteworthy that BamA, an emergent target in recent years, mainly comprises the βbarrel folding complex (BAM),
which serves as the essential chaperone and translocator, folding the outer membrane proteins of Gramnegative
bacteria.
14
The designed chimeric peptidomimetic antibiotics harboring a βhairpin peptide macrocycle, targeting
both LPS and BamA, also achieved considerable success.
15
Undoubtedly, the discoveries of these novel membrane targets and their ligands not only provide a paradigm
for the development of novel antimicrobial drugs but also reveal the feasibility and importance of bacterial cell
membranes as potential antibacterial targets. In bacteria, electron transfer and proton pumping via respiratory chain
on the cytoplasmic membrane generate a proton electrochemical gradient called proton motive force (PMF), which
is considered critical for various cell membrane functions.
16
The dysfunction of PMF directly hinders the synthesis
of adenosine triphosphate (ATP), a kind of unstable highenergy compound that broadly acts as an activator,
YANG ET AL.
|
1069
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
inhibitor, product, or substrate in the complex bacterial physiological network.
17
Generally, the dynamic synthesis
and consumption of ATP is carried out in parallel with many intracellular processes such as signal transduction,
stress response, active transport, polypeptide folding, subunit aggregation, and protein phosphorylation.
1823
Despite the successive findings of potent antibacterial agents targeting PMF, including tridecaptin A1, bacaucin1,
halicin, WRK12, αmangostin (AMG), and isobavachalcone (IBC),
2428
the potential roles of PMF in bacterial
resistance control remain poorly understood. Such a situation warrants a comprehensive investigation of the
function and measurement of PMF and its potential as a novel target in tackling AMR crisis.
This review presents an overview of the cellular functions and characterizations of PMF in bacteria. We
summarize recent advances in developing new antibacterial compounds targeting PMF. Additionally, the adjuvant
potential of bacterial PMF regulators in coordination with existing antibiotics against drugresistant bacteria is
highlighted. Moreover, we explore the potential of PMF disruptors in tackling the transmission of antibiotic
resistance genes (ARGs). Lastly, challenges and future perspectives for the translations and applications of
PMFtargeting compounds are also discussed.
2|FUNCTIONS AND CHARACTERIZATIONS OF PMF IN BACTERIA
PMF, namely the electrochemical potential difference of proton across the cell membrane, comprises two
components: electric potential (ΔΨ) and transmembrane proton gradient (ΔpH).
29
PMF generation in eukaryotes
involves many intimate elements, including mitochondrion, respiratory chain, and ATPase. In the whole biological
system, most energy originated from numerous nutrient transfers from the substrates in the form of highenergy
electrons by nicotinamide adenine dinucleotide (NAD) and flavin adenine dinucleotide (FAD). Subsequently, after
passing through a series of electric mediators in electron transfer chain (ETC), these electrons carried by NADH and
FADH
2
are finally transported to O
2
at the mitochondrial inner membrane (Figure 1A). Meanwhile, the considerable
energy produced along with the electron translocation is used for the transmembrane pumping of protons. More
specifically, protons are transported from the mitochondrial matrix to the intramembranous space, thus reversing
the concentration gradient and creating the electrochemical gradient (PMF) on both sides of the inner membrane.
30
Subsequently, the famous theory called the chemiosmotic hypothesisclaims the coupling of PMF and ATP
synthesis. Driven by the concentration gradient, H
+
flows back to the mitochondrial matrix through specific pores
or channels in F1FoATPase, through which the released energy drives the ATPase to catalyze the interaction of
ADP and Pi to generate ATP.
31
While once the respiration process is interrupted, ATP would be hydrolyzed to
sustain PMF.
Lack of mitochondria in prokaryotes like bacteria leads to the harboring of the respiratory chain and ATPase on
the cytoplasmic membrane. Moreover, the flow of electrons and protons is dynamically changing across the
membrane. Generally, bacteria have a more dedicated and flexible network for electron transport,
32
including
branched pathways and various substitutable terminal electron receptors.
33
Additionally, the respiratory pathways
could be easily adjusted along with the external environment changes. For example, two terminal oxidases in the
aerobic respiratory chain of E. coli, cytochrome o oxidase under the command of cyoABCDE and cydABencoding
cytochrome d oxidase, are converted to cater to the oxygen richness. Herein, only oxygendemanding growth
conditions express cytochrome o oxidase. In contrast, cytochrome d oxidase tends to be less restricted in
oxygen conditions, despite being moderately aerobic. It is also noteworthy that nitrate, fumarate, and
trimethylamine oxide are alternative terminal acceptors when the environment becomes anaerobic.
34
Despite the ability to provide the proximate energy for respiratory ATP synthesis by F1FoATPase, PMF plays
multifaced roles in bacterial bioprocesses, including the import of nutrients, the efflux of toxic products, and flagella
motility (Figure 1B).
16
Substance transport is significant in bacterial biological processes as bacteria undergoing
active metabolism have enormous requirements for a variety of small molecules like ions and macromolecules like
proteins. They primarily serve as energy supplements, raw materials for synthesizing new cellular substances, and
1070
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
intermediate metabolites indispensable to maintaining essential survival and reproductive processes. Meanwhile,
the accumulation of metabolic waste and toxic products causes damage and burden to bacterial cells. Thus, PMF
dependent transmembrane processes with aid of the electrochemical gradient allow the active transport through
the inward and outward pumping of H
+
. Furthermore, H
+
homeostasis is the fundamental premise of all life
activities as enzymes playing catalytic roles require suitable pH conditions. Also, the rotation of bacterial flagella
relies heavily on the energy released from ATP hydrolysis and participates in many physiological activities such as
motion, surface adherence, biofilm formation, and host tissue colonization.
35
Altogether, stable PMF should be
ensured for maintaining healthy bacterial activities.
PMF is distributed in ΔΨ and ΔpH, with stored energy in ΔpH exploiting a persistent change in pH in one or
more cellular compartments. The physical distance between the redox factors and cofactors determines the
influence of ΔΨ on the chargeseparated state. In most cases, PMF remains stable as mutual compensation and
transformation exist between its two components.
36
Specifically, the dissipation of ΔΨ increases the ΔpH to
equilibrate and vice versa.
37
As a testing index, a hydrophobic fluorescent probe 3,3ʹdipropylthiacarbocyanine
iodide (DiSC
3
(5)) is used to probe the dissipation of ΔΨ (Figure 2A).
38
DiSC
3
(5) is a kind of membranelabeling
fluorescence probe, which is sensitive to small changes of membrane potential (ΔΨ) or membrane configuration.
During cell membrane depolarization or permeabilization, the transmembrane electric gradient is disrupted.
Therefore, this probe is translocated to the outer membrane space with enhanced fluorescence units. In contrast,
hyperpolarization of the cell membrane or the loss of chemical gradient (ΔpH) enhances ΔΨ. DiSC
3
(5) uptake
activities will be more significant in this case, weakening the related fluorescence intensity. Together, the increase
of fluorescence suggests the dissipation of ΔΨ, while a decrease otherwise implies the disruption of ΔpH
(Figure 2B). Meanwhile, the investigation of H
+
and K
+
flow using specific fluorescent dyes provides more details for
understanding PMF perturbation (Figure 2C). For some classical PMF disrupters such as CCCP (carbonyl cyanide
FIGURE 1 Generation of proton motive force (PMF) and its functions in multiple bacterial intracellular
activities. (A) Elaborate depiction of how the electron transmitters in the respiratory chain complex pump H
+
from
bacterial cytoplasm to the outside. H
+
on the outer side of the membrane cannot return to the inner side freely
through the inner membrane. In this electron transfer process, the proton concentration gradient and membrane
potential difference are established on both sides of the inner membrane, constituting the transmembrane
electrochemical potential gradient of proton, the PMF. (B) Cellular functions of bacterial PMF. Bacterial PMF drives
many cellular processes, including ATP synthesis, transport of molecules by either symport or antiport (e.g., uptake
of nutrients and efflux of toxic drugs), and rotation of bacterial flagella. ΔΨ, electric potential; ΔpH, transmembrane
proton gradient; ATP, adenosine triphosphate; FAD, flavin adenine dinucleotide; NAD, nicotinamide adenine
di·nucleotide. [Color figure can be viewed at wileyonlinelibrary.com]
YANG ET AL.
|
1071
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
mchlorophenylhydrazone) and nigericin, the internal pH is decreased, with a drop in the intracellular potassium
levels in the meantime.
39
This process differs from the action of valinomycin, as only K
+
influx is observed.
Additionally, biocide triclosan (TCL) and antimalarial drug proguanil hydrochloride (PROG) display another
possibility of inducing the enhanced influx of both H
+
and K
+
.
3|PMF AS A PROMISING TARGET FOR NOVEL ANTIMICROBIAL
AGENTS
As increasing therapeutic drugs gradually lose their efficacies against drugresistant pathogens, developing novel
antimicrobial agents with distinct targets is urgently warranted. As mentioned above, PMF regulates many critical
physiological processes, such as ATP synthesis, signal transduction, macromolecule synthesis, and proteostasis in
bacterial cells.
31
As a result, those compounds with PMFdisturbing activities are potential candidates for
suppressing bacterial infections, especially involving transmembrane transport of solutes or cytoplasmic pH
maintenance.
3.1 |Antimicrobial agents acting upon the Δψ component of PMF
The two components of PMF are closely linked as the positively charged protons generate Δψ and the chemical
separation across the membrane otherwise produces the ΔpH. More generally, Δψ contributes more to PMF
FIGURE 2 Measurement of bacterial PMF and ion changes using fluorescent probes. (A) Structural formula of
DiSC
3
(5), a fluorescence dye sensitive to membrane potential. (B) The relationship between the fluorescence
intensity of DiSC
3
(5) and the disruption of two components of the bacterial PMF. (C) Effect of some PMF disruptors
on the influx and efflux of H
+
and K
+
ions. Nigericin decreases intracellular pH and potassium levels, triclosan
decreases internal pH but enhances K
+
influx, while valinomycin only leads to the accumulation of intracellular
K
+
.ΔΨ, electric potential; ΔpH, transmembrane proton gradient; DiSC
3
(5), 3,3ʹdipropylthiacarbocyanine
iodide; PMF, proton motive force. [Color figure can be viewed at wileyonlinelibrary.com]
1072
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
generation than ΔpH due to the production of larger potential energy from charge separation.
31
Suitable membrane
potential is vital for bacterial survival and various activities; meanwhile, bacteria can harbor completely independent
space isolated from the external environment, especially in extreme conditions.
40
Hence, minor ionic disturbances
induced by capable compounds is prone to disturb PMF homeostasis in bacteria, and more severely, its survival.
Thus, the Δψ component of PMF can be regarded as a potential antibacterial target (Table 1and Figure 3A).
As a proofofconcept, several FDAapproved drugs display their antimicrobial capabilities by disrupting the ΔΨ
component. For example, daptomycin,
41
telavancin,
42
and HT61,
43
all work through potassium or calcium
dependent dissipation of ΔΨ, permeabilizing and depolarizing the cytoplasmic membrane. Moreover, pyrazinamide
(PZA), an effective firstline drug against Mycobacterium tuberculosis, is hydrolyzed to pyrazinoic acid (POA) after
undergoing a series of intraand extracellular processes, which leads to the intracellular accumulation of protons
and the loss of membrane potential.
44
Additionally, some natural products derived from microorganisms or other
sources have been reported to dissipate bacterial ΔΨ. Examples include Lacticin 3147, a bacteriocin produced by
Lactococcus lactis subsp. lactis DPC3147 against a broad range of Grampositive bacteria.
45
At low concentrations,
PMF promotes the interaction between lacticin 3147 and the cytoplasmic membrane, producing the K
+
selective
pores and immediate membrane potential dissipation. Also, Farha et al. performed a highthroughput screen to
identify molecules that specifically dissipate PMF in S. aureus.
46
As a consequence, molecules I1I3 are proved to be
TABLE 1 Antimicrobials acting upon the ΔΨ component of bacterial proton motive force.
Compounds Antibacterial spectrum MIC values References
Daptomycin S. aureus 8μg/mL [41]
Telavancin Grampositive bacteria 0.06 μg/mL [42]
HT61 S. aureus 8μg/mL [43]
Pyrazinamide M. tuberculosis 50 μg/mL [44]
Lactincin 3147 Grampositive bacteria / [45]
Molecule I1I3 S. aureus 28μg/mL [46]
JBC 1847 S. aureus 0.52μg/mL [47]
Oritavancin S. aureus 0.54μg/mL [48]
SC5005 S. aureus 0.5 mg/L [49]
Blestriacin S. aureus 28 µg/mL [50]
Pyrazolopyrimidinnons S. aureus 540 μM[51]
IMRG4 S. aureus 2550 mg/L [52]
Paenipeptin C' P. aeruginosa 12μg/mL [53]
S. aureus 1632 μg/mL
PalαMSH (613) MRSA >45.45 μM[54]
PalαMSH (1113) 11.36 μM
WRK12 MDR Gramnegative bacteria 2 μg/mL [24]
WW307 MDR bacteria 18μg/mL [55]
Bacaucin1 MRSA 4 μg/mL [27]
Abbreviations: ΔΨ, electric potential; MDR, multidrugresistant; MIC, minimum inhibitory concentration; MRSA, methicillin
resistant Staphylococcus aureus; M. tuberculosis, Mycobacterium tuberculosis; P. aeruginosa, Pseudomonas aeruginosa;
S. aureus, Staphylococcus aureus.
YANG ET AL.
|
1073
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
the potential modulators of ΔΨ, subsequently hindering electron transport and ATP synthesis. In particular,
molecule I1 is identified as a type II topoisomerase inhibitor in E. coli, and molecule I3 is a 7,9dialkylpurinium salt
naturally isolated from marine sponges as a secondary metabolite.
56,57
Furthermore, the emergence of some synthesized compounds aimed at disrupting ΔΨ is also exciting. JBC
1847, a phenothiazine derivative, is a positively charged cationic amphiphilic compound exhibiting significantly
improved antistaphylococcal activity through membrane depolarization.
47
Likewise, oritavancin is a semisynthetic
lipoglycopeptide effective in eliminating S. aureus cells in an exponential growth state.
48
The underlying
mechanisms include cell wall injury and membrane potential disruption. In our previous studies, some
synthetic antimicrobial peptides (AMPs) are also exploited to have antibacterial activities by disrupting ΔΨ.
FIGURE 3 Chemical structures of representative antibacterial compounds as (A) ΔΨ disruptors and
(B) ΔpH disruptors in fighting against Grampositive bacteria or Gramnegative bacteria. ΔΨ,electric
potential; ΔpH, transmembrane proton gradient; AMG, αmangostin; IBC, isobavachalcone. [Color figure can
be viewed at wileyonlinelibrary.com]
1074
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Specifically, an amphiphilic peptide WRK12 is designed to cope with various multidrugresistant (MDR) bacteria,
including methicillinresistant S. aureus (MRSA), mcr1positive E. coli (MCRPEC), and tigecyclineresistant E. coli
through membrane permeabilization, ΔΨ dissipation, and reactive oxygen species (ROS) accumulation.
24
Similarly,
in combating MDR pathogens, WW307 peptide damages the membrane by selectively targeting multiple
components such as LPS in the outer membrane and PG in the bacterial cytoplasmic membrane,
55
disrupting ΔΨ
and enhancing ROS. Additionally, the safer derivative of bacaucin, ringopened heptapeptide bacaucin1, shows
specific antibacterial activity against MRSA by dissipating the ΔΨ component,
27
which is similar to lysocin.
12
Recently, more compounds such as sorafenib derivative SC5005,
49
blestriacin,
50
pyrazolopyrimidinones,
51
IMRG4,
52
paenipeptin C(C8Pat),
53
PalαMSH (613), and PalαMSH (1113)
54
have been identified as
potential ΔΨ dissipators to fight against various notorious pathogenic bacteria.
3.2 |Antimicrobial agents acting upon the ΔpH component of PMF
As another component of PMF, ΔpH also plays an integral part in many membrane processes. The ΔpH directly
influences PMF. Generally, the antibacterial activity of the potential dissipaters of ΔΨ is intensely enhanced when
the pH becomes alkaline, decreasing the pH gradient. In contrast, a shift toward acidity increases ΔpH, reinforcing
the antibacterial ability of the ΔpH dissipators. The representative ΔpH dissipators are listed in Table 2and
Figure 3B.
In this context, some membranetargeting peptide compounds show tremendous potential. As a nonribosomal
peptide, tridecaptin A1 originated from Bacillus and Paenibacillus species is effective against various Gramnegative
bacteria, including MDR Klebsiella pneumoniae and E. coli.
28
Independent of membrane lysis or depolarization,
tridecaptin A1 causes pore formation and the loss of proton gradient on the inner membrane by specifically binding
TABLE 2 Antimicrobials acting upon the ΔpH component of bacterial proton motive force.
Compounds Antibacterial spectrum MIC values References
Tridecaptin A1 Gramnegative bacteria 3.1350 μg/mL [28]
EcDBS1R4 E. coli 25 μg/mL [58]
Hepcidin 20 and 25 E. coli 25/50 μg/mL [59]
Bifidobacterium breve (YH68) C. difficile /[60]
Bile salts S. aureus 20/1/>200 mM [61]
Nisin L. monocytogenes /[62]
Halicin M. tuberculosis 2 µg/mL [26]
A. baumannii
Molecule D1D3 S. aureus 8128 μg/mL [46]
Niclosamide H. pylori 0.25 µg/mL [63]
αmangostin (AMG) Isobavachalcone (IBC) MRSA 0.5 µg/mL [25]
4 µg/mL
Nature flavones MRSA 18 µg/mL [64]
Abbreviations: ΔpH, transmembrane proton gradient; A. baumannii, Acinetobacter baumannii; C. difficile, Clostridioides
difficile; E. coli, Escherichia coli; H. pylori, Helicobacter pylori; L. monocytogenes, Listeria monocytogenes; MIC, minimum
inhibitory concentration; MRSA, methicillinresistant Staphylococcus aureus; M. tuberculosis, Mycobacterium tuberculosis; S.
aureus, Staphylococcus aureus.
YANG ET AL.
|
1075
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
with lipid II in Grampositive bacteria. Likewise, an AMP, EcDBS1R4, causes membrane hyperpolarization and the
increased assembly of inner membranelike (IML) vesicles in E. coli.
58
Herein, rearrangements of CL, a constitutive
phospholipid closely contacting with electron transport chain proteins, block the dissipation of proton potential
used for ATP synthesis, eventually causing hyperpolarization and the subsequent collapse of ΔpH. Additionally,
another research discusses the pHdependent interference of E. coli membranes by human AMPs hepcidin 20
and 25.
59
Biological agents such as probiotics and organic salts are widely used daily. Some of them harbor impressive
antibacterial properties by targeting ΔpH. For example, bifidobacterium breve (YH68) is common in food
fermentation. It can counter Clostridioides difficile infections by disrupting ΔΨ and ΔpH of the cytoplasmic
membrane, leading to pore formation and cell disintegration.
60
Bile salt supplements lower cholesterol levels by
emulsifying lipids, providing a new train of thought for controlling drugresistant bacteria.
65
It has been reported
that both conjugated and unconjugated bile salts at subinhibitory concentrations collapse the bioenergetic process
by reducing intracellular pH in S. aureus.
61
Moreover, food preservatives such as bacteriocin produced by
Lactobacillus spp. effectively eliminates Listeria monocytogenes cells.
62
With 1 μg/mL nisin, a ribosomally
synthesized peptide commonly used as a food preservative, entirely dissipates ΔpH in a timeand
concentrationdependent manner; however, ΔΨ is only slightly decreased. Also, the natural flavones isolated
from the Morus alba (white mulberry), such as kuwanon G, kuwanon H, mulberrin, and morusin, effectively kill some
MRSA isolates by collapsing ΔpH.
64
Likewise, AMG and IBC bind to the membrane phospholipids and dissipate
ΔpH, leading to metabolic perturbation.
25,66
In the face of the increasing emergence and dissemination of MDR
bacterial pathogens, these natural products have immense potential to provide new chemical scaffolds, thus
accelerating the discovery of antibacterial agents.
Recently, a deep neural network is trained to predict molecules with antimicrobial activities.
26
Growth
inhibition is an essential index in library screening. A novel antibiotic termed halicin stands ahead with
predicted antimicrobial activity, lower minimum inhibitory concentration (MIC), and lower structural similarity
to its nearest neighbor antibiotic. Most inspiringly, halicin effectively copes with antibiotictolerant E. coli and
M. tuberculosis and inhibits the growth of numerous MDR Gramnegative clinical isolates, demonstrating its
unusual antimicrobial effect. Moreover, wholetranscriptome sequencing comprehensively explains how
halicin combines with iron to cause the dissipation of transmembrane ΔpH. It is an opportune time for
antibiotic discovery based on machine learning approaches. The expectation for narrowspectrum agents and
new scaffolds circumventing preexisting resistance determinants can be gradually achieved with decreasing
costs. Furthermore, considering the compensation mechanism between ΔΨ and ΔpH and their sensitivities to
a series of perturbations, a more targeted highthroughput screen is performed and molecules D1D3 with
capabilities to collapse transmembrane pH gradient of S. aureus are found.
46
Discovering new effective agents
is a stunning process, while repurposing conventional drugs achieves more productivity. Niclosamide
effectively treats cestodes (tapeworms) by inhibiting energy derived from anaerobic metabolism.
67
It is also an
anthelmintic drug and has been listed as an essential medicine by World Health Organization (WHO).
68
A recent study has further revealed its activity against Helicobacter pylori via disruption of transmembrane pH,
especially at lower MIC values.
63
For a long time, membraneactive agents have been set aside during the process of developing novel
antimicrobials as their propensity to induce toxicity in mammals.
69
However, considering its performance in treating
drugresistant bacteria and the recent success of some antibiotic mechanisms, this initially apparent dismal
antibacterial target has regained increasing attention.
7072
As the two components of PMF, ΔΨ and ΔpH, are
compensated mutually for either dissipation. This phenomenon enables PMF to be exquisitely susceptible to a
series of perturbances, rendering them fatal for bacteria.
73
Collectively, we highlight the significance of ΔΨ and
ΔpH in all bacterial activities, leading to the possibility of discovering previously unknown compounds as new
antimicrobial agents.
1076
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4|ADJUVANT POTENTIAL OF BACTERIAL PMFACTING COMPOUNDS
Although the discovery of novel antimicrobial agents is the most direct approach to killing these drugresistant
pathogens, the very low outputtoinput ratio including substantial economic and time investment, coupled with the
rapid generation of resistant bacteria, limits the feasibility of this strategy. In contrast, restoring the efficacy of
initially effective antimicrobials using antibiotic adjuvants offers an innovative approach to minimizing the
emergence and impact of antibiotic resistance from a novel perspective.
Generally, the resistance mechanism has been comprehensively illustrated until now. Besides inherent
resistance, acquired resistance can be attributed to four main reasons: producing modifying enzymes, altering
targets, active efflux, or reduced uptake.
74
The latter two are closely related to the functions of bacterial
membrane, especially those of PMF. In the bacterial respiratory chain, PMF can serve as an acute energy responder
for bacterial metabolism and vitality.
75
Also, relying on the electrochemical gradient across the membrane, PMF
undoubtedly controls the influx and efflux, which is essential for intracellular antibiotic accumulation.
76
Altogether,
it is not hard to surmise the potential roles of PMF in developing resistance or even tolerance, which can be
attentively considered in the deployment of new antibiotic adjuvants. In Table 3, we provide a list of these
representative PMFtargeting adjuvants.
4.1 |PMF correlates with bacterial resistance and tolerance
It is indisputable that we still get confronted with the intensified resistance crisis despite the numerous antibiotic
categories, warranting urgent novel antibacterial strategies from different angles. Recently, bacterial metabolism
has been implicated in the development of resistance.
85
Studies have thoroughly profiled drug resistance issues
across the whole metabolic state of bacteria, and explored ways to increase intracellular antibiotic concentrations
and reverse antibiotics resistance by establishing overall metabolism regulation.
86
Their efforts identified some key
metabolites that rearrange resistant metabolome to a sensitive, promoting the uptake of antibiotics and effectively
diminishing resistant bacteria.
77,87
Moreover, central carbon energy metabolismPMF is discovered as a new
resistance metabolism regulation pathway.
78,88
These findings not only highlight the critical reason behind the
decreased PMF in resistant bacteria, but also connect the tricarboxylic acid (TCA) cycle, glycolysis, and pyruvate
metabolism with the respiratory chain and PMF. This creates a new therapeutic method that combines antibiotics
and metabolism regulators. Furthermore, drugresistant bacteria have active efflux tactics to decrease intracellular
antibiotic concentration. Therefore, some compounds successfully perturb membrane PMF and damage the normal
operation of efflux pumps and ATP synthesis, ultimately restoring bacterial susceptibility to therapeutic drugs.
80
Tolerance describes how nongrowing or slowgrowing bacteria survive bactericidal antibiotics that target an
active metabolic state. The nongrowing phenotype subpopulation occupying a small fraction of the bacterial
population, such as that in biofilms, is called persisters.
89
Recent studies have shown that intermittent antibiotic
exposure promotes rapid evolution of tolerance and persistence.
90,91
Moreover, tolerance contributes to resistance
evolution by giving rise to numerous possible mutations in a bacterial population under periodic exposure to high
antibiotic concentrations.
92
As a consequence, blocking the development of tolerance makes a significant
difference in resistance evolution. Thus, the factors underlying tolerance phenotype are worthy of being studied
intensively. To investigate the physiological responses outside metabolism shutdown during tolerance evolvement,
Wang et al. showed that active efflux and the activities of some critical membrane proteins due to PMF contribute
to conferring the starvationinduced tolerance phenotype in E. coli.
93
Meanwhile, disrupting PMF through
ionophore CCCP or inhibitors of ETC complex otherwise suspends this phenotype. It has been shown that the
preaddition of mannitol to lung infections of Pseudomonas aeruginosa persister cells during biofilm growth reverts
the persister phenotype in a PMFdependent manner.
94
Collectively, both tolerant and persistent phenotypes have
a close link with the state of PMF, suggesting a potential biological switch to block the pathway to resistance.
YANG ET AL.
|
1077
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 3 Antibiotic adjuvants targeting bacterial proton motive force and their mechanisms of action.
Compounds Antibiotics Mechanisms References
Glycine, serine, and threonine Kanamycin ΔΨ[77]
Amino acids biosynthesis
TCA cycle
Fructose Kanamycin ΔΨ[78]
TCA cycle
Llysine Kanamycin ΔpH[79]
/Gentamicin Antibiotic uptake
/Amikacin
Pyruvate, succinate and glutamate Tobramycin ΔpH[75]
Biofilm inhibitory activity
Antibiotic uptake
ML7 Tigecycline ΔpH,ΔΨ[80]
Efflux pump
ATP synthesis
Carprofen Doxycycline ΔpH[81]
Efflux pump
Protein synthesis
Benzydamine Doxycycline ΔpHΔΨ[76]
Antibiotic uptake
Protein synthesis
Efflux pump
Metformin Doxycycline/Minocycline ΔpH,ΔΨ[82]
Antibiotic uptake
Efflux pump
TetA protein synthesis
Citral Norfloxacin ΔΨ[83]
Efflux pump
IMRG4 Norfloxacin ΔΨ[52]
Membrane permeabilization
Efflux pump (NorA)
Melatonin Colistin ΔΨ[84]
Efflux pumps
Protein synthesis
Abbreviations: ΔΨ,electricpotential;ΔpH, transmembrane proton gradient; ATP, adenosine triphosphate; TCA,
tricarboxylic acid.
1078
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Herein, we are about to elaborate on the adjuvant functions of PMF regulators in two main classes of antibiotics, as
their primary modes of action rely on different components.
4.2 |PMF in the development of aminoglycoside adjuvants
Aminoglycoside antibiotics exert solid antibacterial activities against the broad extension of notorious MDR bacteria
such as P. aeruginosa, S. aureus, and E. coli.
95
They are mainly composed of a 2deoxystreptamine (2DOS) ring and
several aminomodified sugars, usually connected by glycosidic bonds to form glycosides.
96
Generally,
aminoglycosides interact with the bacterial outer membrane and arrive at the protein synthesis site in an
energydependent way, irreversibly binding to the 30 S subunit of the 16 S ribosomal RNA (rRNA).
97
The following
consequences include codon misreading, interrupted peptide elongation, and even the production of malfunction-
ing protein, disrupting the structure of the cell membrane and increasing intracellular antibiotic accumulation.
98
Representative members like streptomycin, gentamicin, neomycin, and paromomycin are usually suggested in
treating clinical infections such as tuberculosis, brucellosis, tularemia, enterococcal endocarditis, and hepatic
encephalopathy.
99
Aminoglycoside antibiotics are gradually getting attentioninclinicaluse,withtheresistanceofβlactam
antibiotics and fluoroquinolones in Gramnegative bacteria becoming severe.
100
Despite this trend, controlling
toxicity and hindering resistance are always of foremost importance. In the treatment of MDR Edwardsiella
piscicida, the combinations of glycine, serine, and threonine present the best efficacy, leading to increased
pyruvate dehydrogenase, αketoglutarate dehydrogenase, and succinate dehydrogenase activities in TCA cycle.
This line of treatment is also marked by enhanced membrane potential and intracellular kanamycin.
77
Similarly,
fructose effectively activates the TCA cycle to produce NADH, generating PMF to increase the uptake of
kanamycin in treating MDR Edwardsiella tarda.
78
Later, Webster et al. highlighted the relationship between
antibiotic toxicity, ROS production, and PMF, elaborating how the uncoupler 2,4dinitrophenol (DNP) weakens
the efficacy of gentamicin through ΔΨ dissipation in pathogenic E. coli.
66
This conversely indicates that PMF
actively underpins respirationmediated potentiation of aminoglycoside lethality. These findings strongly
suggest that the main adjuvants of aminoglycosides enhance antibiotic efficacy by boosting ΔΨ to regulate
metabolism and activate respiration (Figure 4A). Furthermore, the exogenous addition of Llysine potentiates
aminoglycoside activity against Acinetobacter baumannii by enhancing ΔpH and increasing antibiotic uptake.
79
Endogenous metabolites secreted from 3D lung cells also enhance the biofilm inhibitory activity of tobramycin
and promote antibiotic uptake against the opportunistic pathogen P. aeruginosa through the same
mechanism.
75
These two cases improve our understanding of the underlying modes of action of aminoglycoside
adjuvants on bacterial PMF.
4.3 |PMF in the development of tetracyclines adjuvants
Tetracycline antibiotics have broadspectrum antimicrobial activities against microorganisms and serve as the most
widely used antibiotic in veterinary clinics worldwide.
101,102
Numerous compounds are subsequently included in
this antibiotic category, such as the natural product chlortetracycline and second and thirdgeneration derivatives
with improved antibacterial potency and resistance coverage. Typical drugs are semisynthetic rolitetracycline,
doxycycline, minocycline, and, more importantly, tigecycline.
103
Nowadays, tigecycline is one of the lastresort
antibiotics to treat complicated infections caused by MDR bacteria, including vancomycinresistant Enterococcus
faecium, MRSA and carbapenemresistant Enterobacteriaceae (CRE).
104
Unfortunately, due to increasing clinical
usage, tetracycline resistance is conferred in susceptible bacterial populations through horizontal gene transfer
(HGT), triggering overexpression of efflux pumps and ribosome protection.
105107
Altogether, it is urgent to develop
YANG ET AL.
|
1079
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
potent adjuvants for tigecycline or other tetracyclines. Sun et al. found that tigecycline with ML7 entirely inhibits
bacterial growth with synergistic bactericidal activities against drugresistant isolates.
80
The underlying mechanisms
include the increased ΔpH and the subsequent inhibition of the efflux pump and ATP synthesis. Moreover,
coexposure to carprofen restores doxycycline susceptibility in methicillinresistant Staphylococcus pseudintermedius
(MRSP) ST71.
81
Herein, TetKmediated drug efflux is paralyzed by ΔpH dissipation and the intracellular protein
synthesis is then broken off. Our previous studies showed that nonantibiotic pharmaceutical benzydamine is also
effective against a series of clinically hazardous pathogens in combination with tetracyclines,
76,108
including
TMexCDTOprJexpressing bacteria that confer highlevel tigecycline resistance.
109
Indepth studies indicate that
the potentiation of benzydamine to tigecycline correlates with PMF changes. On the one hand, benzydamine
addition disrupts ΔΨ and upregulates ΔpH to compensate, leading to the increased uptake of tetracyclines and the
inhibition of protein synthesis. On the other hand, dissipated PMF reduces efflux pump activity, contributing to the
increased intracellular drug accumulation, thus more effectively killing bacteria. Similarly, the adjuvant potential of
PMFacting drugs is also thoroughly clarified in the case of metformin, a firstline antidiabetic drug, which could be
a potent adjuvant for tetracyclines in the fight against various MDR pathogens, including the notorious MRSA.
82
The increased ΔpH promotes antibiotic uptake, while the decrease of PMF inhibits the activity of the efflux pump
and the protein synthesis of TetA. Altogether, ΔpH critically regulates tetracyclines resistance and can serve as a
FIGURE 4 Adjuvants of aminoglycoside or tetracycline antibiotics act by targeting different components of
bacterial PMF. (A) Active bacterial metabolism induced by exogenously adding adjuvants makes aminoglycosides
more effective as they potentiate the tricarboxylic acid (TCA) cycle and produce more NADH, which together
boosts ΔΨ and PMF, thus promoting aminoglycoside uptake. (B) Tetracyclines adjuvants not only increase ΔpH to
enhance intracellular drug accumulation but also disrupt PMF to paralyze the function of the efflux pump, thereby
restoring the antibacterial activity of tetracyclines. ΔΨ, electric potential; ΔpH, transmembrane proton gradient;
ATP, adenosine triphosphate; PMF, proton motive force. [Color figure can be viewed at wileyonlinelibrary.com]
1080
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
promising target for screening effective tetracycline adjuvants, significantly improving drug uptake and reducing
efflux activity (Figure 4B).
4.4 |PMFmodulating compounds resensitize other antibiotic classes
Besides these two classical categories, PMF also restores the susceptibility of resistant bacteria to many other
antibiotics. Fluoroquinolones harbor a broader antibacterial spectrum and better efficacy than quinolones due to
the additional fluorine atom at the sixth position.
110
Owing to such properties, the current UK prescribing
guidelines once recommended fluoroquinolones as secondline agents to maintain their effectiveness. However,
resistance development has never slowed, warranting suitable sensitizers.
111
Previous research has reported high
level fluoroquinolone resistance of Shigella dysenteriae induced by a PMFdependent efflux system.
112
Several
fluoroquinolone adjuvants are found to influence ΔΨ or ΔpH. For example, in the battle against MRSA, one
monoterpenoid aldehyde citral exerts a synergistic effect with norfloxacin through multiple mechanisms, including
efflux pump inhibition and membrane potential disruption.
83
Similarly, a novel bifunctional chalcone IMRG4
effectively inhibits MRSA by potentiating the activity of fluoroquinolone through membrane depolarization and
permeabilization.
52
Additionally, colistin is a last resortantibiotic for treating infections caused by many dangerous Gram
negative pathogens. Some previous studies in Burkholderia thailandensis have demonstrated a DedA family protein
as a transmembrane transporter in the development of colistin resistance via covalent modification of lipid A in
LPS.
113
However, the activity of DbcA (DedA of Burkholderia required for colistin resistance) is protondependent,
with PMF disruptors restoring colistin efficacy.
114
Additionally, our previous study illustrated how melatonin
reverses MCRmediated colistin resistance in Gramnegative bacteria by disrupting membrane potential and
inhibiting efflux pumps and ABC transporter.
84
Meanwhile, the uptake of erythromycin and azithromycin,
representatives of macrolides in Haemophilus influenzae ATCC 19418, is evidently enhanced through a passive
diffusion upon ΔpH disruption.
115
Taken together, these successful examples encouragingly demonstrate the
universal applicability of bacterial PMF in regulating bacterial resistance. The recent decades have witnessed
the diminishing effectiveness of various originally potent antibiotics, posing a global threat to public health. In this
regard, exploiting new classes of antibiotics is timeconsuming and requires a long cycle while restoring the
susceptibility of existing antibiotics has more advantages. PMF modulators display their adjuvant potential against
resistance as sensitizers of antimicrobials and can reverse the resistance or tolerance phenotype. Such findings
immensely aid this branch of research in the present and the future, finding the optimal antibacterial strategies with
the lowest possibility of inducing resistance. The involvement of bacterial PMF is opening new insights toward
identifying reliable antibiotic adjuvants. We anticipate the development of similar combinations in treating
nosocomial infections.
5|PMF DISRUPTORS PREVENT THE TRANSMISSION OF ARGs
Compared with intrinsic resistance, acquired resistance is recognized as the leading cause of the rapid prevalence of
drugresistant bacteria worldwide.
74,116118
ARGs on the chromosome are usually replicated during bacterial
division and proliferation. At the same time, plasmidmediated HGT is achieved through conjugation,
transformation, transduction, and vesiduction.
119
Direct transmission through conjugation and indirect transmission
through transformation have a subversive impact on the whole genetic environment in the bacterial population.
120
To effectively control the propagation of AMR, it is necessary to suppress these pathways through additional
interferences. Thus, preventing the plasmidmediated horizontal transfer of ARGs provides a feasible approach to
minimizing this global crisis.
YANG ET AL.
|
1081
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
HGT involves the transmembrane process and requires energy consumption, indicating the potential role of
PMF in its regulation. In agreement with this notion, increased PMF is reported to provide more power for
exogenous DNA uptake in many conjugation processes.
121,122
Interestingly, we recently found that melatonin
effectively inhibits the horizontal transfer of mcrharboring plasmids between intraand interspecies (Figure 5A).
123
We also highlight how melatonin disrupts bacterial PMF, which is intimately related to the energy supply and
conjugative transfer process. These findings shed new light on screening effective conjugation inhibitors by
perturbing bacterial PMF. Not merely, compounds blocking the competence can also interrupt transformation as
competence provides a physiological state to meet the requirements for the uptake and integration of the
exogenous DNA (Figure 5B).
124
Therefore, Domenech et al. identified COMblockers in Streptococcus pneumoniae
from 1380 drugs and further illustrated their mechanisms of action by disrupting the PMFdependent export
of quorumsensing peptide that activates competence.
39
The following studies confirm the efficacy of these
COMblockers in inhibiting transformation in human cell line colonization and in vivo models. Altogether, it would
be interesting in practice to develop strategies restricting resistance transmission through PMF disruption to reduce
the global burden of AMR.
6|CONCLUSION AND OUTLOOK
AMR is one of the most pressing issues faced by the healthcare system today. Such a situation demands the urgent
development of novel antibacterial agents with distinct scaffolds or targets to combat the resistance crisis in the
postantibiotic era. In the early stages, all kinds of antibiotics are subsequently discovered with targets in the cell
walls, on the membrane or during the synthesis of protein and genetic material.
74
Yet, in recent years, researchers
have tried to steer clear from discovering membraneactive agents either damaging the membrane integrity or
FIGURE 5 The role of PMF in exogenous DNA uptake during bacterial conjugation and transformation. (A) The
potential mechanisms related to PMF in the donor strain during conjugation. For example, melatonin inhibits DNA
uptake, flagellar motility, and ATP synthesis through PMF collapse, especially disrupting ΔΨ. Changes in the
intracellular ion concentrations further support the PMF disruption. (B) The underlying mechanisms of competence
and transformation inhibition in S. pneumoniae. The comCencoded competencestimulating peptide (CSP) is
cleaved and transported outside the cell by ComAB with the support of PMF. Then, histidinekinase ComD is
autophosphorylated upon binding with CSP. The phosphate group is then transferred to the regulator ComE to
form phosphorylated ComE, which activates the former competence genes, including comAB and comCDE,
generating a positive feedback pathway. Meanwhile, comX is also activated to export a sigma factor (SigX),
contributing to the activation of genes responsible for transformation and DNA repair. ΔΨ, electric potential; ΔpH,
transmembrane proton gradient; ATP, adenosine triphosphate; PMF, proton motive force. [Color figure can be
viewed at wileyonlinelibrary.com]
1082
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
targeting energetics, especially those with a propensity to induce mammalian toxicity. Meanwhile, the gradually
emerging combined consensus indicates that potential candidates acting on cell membranes should be excluded
from further studies in wholecell screenings. Analytical methods for detecting these undesirable compounds are
also available to ease this process.
125,126
But recently, PMF has been frequently reported to have indispensable
functions in various membrane processes during the safe administration of potent antimicrobials, vastly raising our
confidence and anticipation.
70,127
This phenomenon implies that this originally unpopular and unexplored drug
target related to the energetic pathways of bacterial membrane is being rejuvenated for further research.
From a comprehensive perspective, we conclude three main approaches to combating infections of pathogenic
bacteria, that is, exploiting direct bactericidal or bacteriostatic drugs, identifying novel antibiotic adjuvants, and
blocking the transmission of AMR. In this review, we have illustrated the roles of bacterial PMF modulators in
regulating these three aspects with multitudinous examples (Figure 6). Most importantly, PMF is closely linked with
the respiratory chain that participates in intracellular energy transformation and transmembrane substance
transport, satisfying the essential physiological requirements and supporting common bacterial activities. Also,
stable membrane potential allows the ion flows on both sides of the membrane to maintain a dynamic balance.
40
Herein, H
+
homeostasis is pivotal to the bacterial inner environment, with a suitable pH ensuring the normal
functions of various critical enzymes. Therefore, such important status and flexible changes render PMF a target
that can be regulated through external intervention. Indeed, we are excited to identify some potent agents showing
direct bactericidal or bacteriostatic effects via PMF interruption (Figure 6A). Moreover, a dedicated bacterial
internal regulation network crucially regulates bacterial survival. The pressure exerted by aminoglycoside antibiotics
FIGURE 6 Three strategies to cope with drugresistant pathogens by targeting bacterial PMF. (A) PMF is a
promising target for new antibacterial agents. (B) The supplementation of PMF regulators restores the antibacterial
activity of certain antibiotics against drugresistant bacteria. (C) PMF disruptors block the propagation of resistance
genes by preventing conjugation and transformation. PMF, proton motive force. [Color figure can be viewed at
wileyonlinelibrary.com]
YANG ET AL.
|
1083
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
causes the bacteria to survive at the cost of reduced metabolism.
85
Main energy circles may have defects in one or
several segments, causing downstream responses such as PMF dissipation and ATP synthesis inhibition. In this way,
the suitable addition of exogenous substances, especially those defective metabolites, accelerates the accumulation
and uptake of aminoglycosides by promoting PMF, primarily by regulating ΔΨ, thus resensitizing drugresistant
bacteria (Figure 6B). Meanwhile, some PMF disruptors serve as potent adjuvants for tetracycline antibiotics,
restoring their antibacterial activities via enhanced uptake due to an increased ΔpH and reduced efflux by inhibiting
PMF. Furthermore, comprehensive consideration reveals that suppressing bacterial resistance is warranted before
it becomes rampant, with more attention being paid to cutting off transmission pathways. Conjugation and
transformation are common ways of plasmidmediated horizontal transfer of ARGs.
120
Recent studies highlight that
disrupted PMF induced by some compounds cannot maintain the competence or sufficiently provide energy for
exogenous DNA uptake, thus enormously preventing the dissemination of ARGs (Figure 6C).
39,123
In summary, we highlight that bacterial PMF essentially regulates resistance emergence, evolution, and
transmission. PMF may be one of the promising targets for developing novel strategies to control the increasing
MDR bacterial infections. However, it is also noteworthy that both PMF disruptors and regulators still warrant a
series of preclinical studies. Many setbacks in using these promising compounds, such as nonspecific cytotoxicity on
mammalian cells or other multimodal activities, are undesirable for drug development. For example, despite being
typical ΔΨ disruptors, the applications of polymyxins (i.e., polymyxin B and colistin) are largely limited by safety
concerns such as acute toxicity and doselimiting nephrotoxicity. Likewise, the development of surotomycin
targeting C. difficile through cellular membrane depolarization is eventually suspended after Phase III studies for
unsatisfactory noninferiority criteria compared with vancomycin and a higher propensity to cause nausea and
headache.
128,129
Despite these obstacles, the introduction of new technologies, such as computerassisted design,
will undoubtedly contribute to the development of safer and more effective antibacterial drugs, as reflected in the
success of novel polymycin derivatives with lower nephrotoxicity and improved therapeutic indices.
130132
Therefore, it is worth anticipating the extraordinary efficacy of PMFtargeting compounds in treating MDR bacterial
infections by effectively avoiding their potential side effects.
ACKNOWLEDGMENTS
This work was supported by the National Key Research and Development Program of China (2021YFD1801000
and 2018YFA0903400), National Natural Science Foundation of China (32222084, 32172907 and 32002331),
Jiangsu Agricultural Science and Technology Innovation Fund (CX(21)2010), A Project Funded by the Priority
Academic Program Development of Jiangsu Higher Education Institutions (PAPD), Young Elite Scientists
Sponsorship Program by CAST (2020QNRC001) and 111 Project D18007.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
DATA AVAILABILITY STATEMENT
The data used to support the findings of this study are available from the corresponding authors upon request.
ORCID
Yuan Liu http://orcid.org/0000-0002-9622-6471
REFERENCES
1. Murray CJ, Ikuta KS, Sharara F, et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic
analysis. Lancet. 2022;399(10325):629655.
2. Theuretzbacher U, Outterson K, Engel A, Karlén A. The global preclinical antibacterial pipeline. Nat Rev Microbiol.
2020;18(5):275285.
1084
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3. Galarion LH, Mohamad M, Alzeyadi Z, Randall CP, O'Neill AJ. A platform for detecting crossresistance in
antibacterial drug discovery. J Antimicrob Chemother. 2021;76(6):14671471.
4. Vila J, MorenoMorales J, BallestéDelpierre C. Current landscape in the discovery of novel antibacterial agents. Clin
Microbiol Infect. 2020;26(5):596603.
5. MacNair CR, Farha MA, SerranoWu MH, et al. Preclinical development of pentamidine analogs identifies a potent
and nontoxic antibiotic adjuvant. ACS Infect Dis. 2022;8(4):768777.
6. Zhang N, Ma S. Recent development of membraneactive molecules as antibacterial agents. Eur J Med Chem.
2019;184:111743.
7. Zhou M, Zheng M, Cai J. Small molecules with membraneactive antibacterial activity. ACS Appl Mater Interfaces.
2020;12(19):2129221299.
8. Pader V, Hakim S, Painter KL, Wigneshweraraj S, Clarke TB, Edwards AM. Staphylococcus aureus inactivates
daptomycin by releasing membrane phospholipids. Nat Microbiol. 2016;2:16194.
9. Song M, Liu Y, Huang X, et al. A broadspectrum antibiotic adjuvant reverses multidrugresistant Gramnegative
pathogens. Nat Microbiol. 2020;5(8):10401050.
10. Johnston CW, Skinnider MA, Dejong CA, et al. Assembly and clustering of natural antibiotics guides target
identification. Nat Chem Biol. 2016;12(4):233239.
11. O'Rourke S, Widdick D, Bibb M. A novel mechanism of immunity controls the onset of cinnamycin biosynthesis in
Streptomyces cinnamoneus DSM 40646. J Ind Microbiol Biotechnol. 2017;44(45):563572.
12. Hamamoto H, Urai M, Ishii K, et al. Lysocin E is a new antibiotic that targets menaquinone in the bacterial membrane.
Nat Chem Biol. 2015;11(2):127133.
13. Kaur H, Jakob RP, Marzinek JK, et al. The antibiotic darobactin mimics a βstrand to inhibit outer membrane
insertase. Nature. 2021;593(7857):125129.
14. Imai Y, Meyer KJ, Iinishi A, et al. A new antibiotic selectively kills Gramnegative pathogens. Nature. 2019;576(7787):
459464.
15. Luther A, Urfer M, Zahn M, et al. Chimeric peptidomimetic antibiotics against Gramnegative bacteria. Nature.
2019;576(7787):452458.
16. Le D, Krasnopeeva E, Sinjab F, Pilizota T, Kim M. Active efflux leads to heterogeneous dissipation of proton motive
force by protonophores in bacteria. mBio. 2021;12(4):e0067621.
17. Zhou J, Liu L, Shi Z, Du G, Chen J. ATP in current biotechnology: regulation, applications and perspectives. Biotech
Adv. 2009;27(1):94101.
18. Yuroff AS, Sabat G, Hickey WJ. Transportermediated uptake of 2chloroand 2hydroxybenzoate by Pseudomonas
huttiensis strain D1. Appl Environ Microbiol. 2003;69(12):74017408.
19. Klipp E, Nordlander B, Krüger R, Gennemark P, Hohmann S. Integrative model of the response of yeast to osmotic
shock. Nature Biotechnol. 2005;23(8):975982.
20. Watanabe Y, Oshima N, Tamai Y. Coexpression of the Na
+
/H
+
antiporterand H
+
ATPase genes of the salttolerant
yeast Zygosaccharomyces rouxii in Saccharomycescerevisiae.FEMS Yeast Res. 2005;5(45):411417.
21. Kragol G, Lovas S, Varadi G, Condie BA, Hoffmann R, Otvos L. The antibacterial peptide pyrrhocoricin inhibits the
ATPase actions of DnaK and prevents chaperoneassisted protein folding. Biochemistry. 2001;40(10):30163026.
22. Deutscher J, Francke C, Postma PW. How phosphotransferase systemrelated protein phosphorylation regulates
carbohydrate metabolism in bacteria. Microbiol Mol Biol Rev. 2006;70(4):9391031.
23. Kipnis Y, Papo N, Haran G, Horovitz A. Concerted ATPinduced allosteric transitions in GroEL facilitate release of
protein substrate domains in an allornone manner. Proc Natl Acad Sci USA. 2007;104(9):31193124.
24. Liu Y, Shi J, Tong Z, Jia Y, Yang K, Wang Z. Potent broadspectrum antibacterial activity of amphiphilic peptides
against multidrugresistant bacteria. Microorganisms. 2020;8(9):1398.
25. Song M, Liu Y, Li T, et al. Plant natural flavonoids against multidrug resistant pathogens. Adv Sci. 2021;
8(15):2100749.
26. Stokes JM, Yang K, Swanson K, et al. A deep learning approach to antibiotic discovery. Cell. 2020;180(4):688702.
27. Liu Y, Ding S, Dietrich R, Märtlbauer E, Zhu K. A biosurfactantinspired heptapeptide with improved specificity to kill
MRSA. Angew Chem Int Ed. 2017;56(6):14861490.
28. Cochrane SA, Findlay B, Bakhtiary A, et al. Antimicrobial lipopeptide tridecaptin A1 selectively binds to Gram
negative lipid II. Proc Natl Acad Sci USA. 2016;113(41):1156111566.
29. Mitchell P. Coupling of phosphorylation to electron and hydrogen transfer by a chemiosmotic type of mechanism.
Nature. 1961;191:144148.
30. Rieger B, Arroum T, Borowski MT, Villalta J, Busch KB. Mitochondrial F1Fo ATP synthase determines the local
proton motive force at cristae rims. EMBO Rep. 2021;22(12):e52727.
31. Berry BJ, Trewin AJ, Amitrano AM, Kim M, Wojtovich AP. Use the protonmotive force: mitochondrial uncoupling
and reactive oxygen species. J Mol Biol. 2018;430(21):38733891.
YANG ET AL.
|
1085
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
32. Kaila VRI, Wikström M. Architecture of bacterial respiratory chains. Nat Rev Microbiol. 2021;19(5):319330.
33. Cotter PA, Chepuri V, Gennis RB, Gunsalus RP. Cytochrome o (cyoABCDE) and d (cydAB) oxidase gene
expression in Escherichia coli is regulated by oxygen, pH, and the fnr gene product. JBacteriol. 1990;172(11):
63336338.
34. Tseng CP, Albrecht J, Gunsalus RP. Effect of microaerophilic cell growth conditions on expression of the aerobic
(cyoABCDE and cydAB) and anaerobic (narGHJI, frdABCD, and dmsABC) respiratory pathway genes in Escherichia
coli.J Bacteriol. 1996;178(4):10941098.
35. Abdulkadieva MM, Sysolyatina EV, Vasilieva EV, et al. Strain specific motility patterns and surface adhesion of
virulent and probiotic Escherichia coli.Sci Rep. 2022;12(1):614.
36. Bakker EP, Mangerich WE. Interconversion of components of the bacterial proton motive force by electrogenic
potassium transport. J Bacteriol. 1981;147(3):820826.
37. Mohiuddin SG, Ghosh S, Kavousi P, Orman MA. Proton motive force inhibitors are detrimental to Methicillin
Resistant Staphylococcus aureus strains. Microbiol Spectr. 2022;10(4):e0202422.
38. Te Winkel JD, Gray DA, Seistrup KH, Hamoen LW, Strahl H. Analysis of antimicrobialtriggered membrane
depolarization using voltage sensitive dyes. Front Cell Dev Biol. 2016;4:29.
39. Domenech A, Brochado AR, Sender V, et al. Proton motive force disruptors block bacterial competence and
horizontal gene transfer. Cell Host Microbe. 2020;27(4):544555.
40. Benarroch JM, Asally M. The microbiologist's guide to membrane potential dynamics. Trends Microbiol. 2020;28(4):
304314.
41. Ma W, Zhang D, Li G, et al. Antibacterial mechanism of daptomycin antibiotic against Staphylococcus aureus based on
a quantitative bacterial proteome analysis. J Proteomics. 2017;150:242251.
42. Al Jalali V, Zeitlinger M. Clinical pharmacokinetics and pharmacodynamics of telavancin compared with the other
glycopeptides. Clin Pharmacokinet. 2018;57(7):797816.
43. Hubbard ATM, Barker R, Rehal R, Vandera KKA, Harvey RD, Coates ARM. Mechanism of action of a membrane
active quinolinebased antimicrobial on natural and model bacterial membranes. Biochemistry. 2017;56(8):
11631174.
44. Lamont EA, Dillon NA, Baughn AD. The bewildering antitubercular action of pyrazinamide. Microbiol Mol Biol Rev.
2020;84(2):e0007019.
45. Ryan A, Patel P, O'Connor PM, Ross RP, Hill C, Hudson SP. Pharmaceutical design of a delivery system for the
bacteriocin lacticin 3147. Drug Delivery Transl Res. 2021;11(4):17351751.
46. Farha MA, Verschoor CP, Bowdish D, Brown ED. Collapsing the proton motive force to identify synergistic
combinations against Staphylococcus aureus.Chem Biol. 2013;20(9):11681178.
47. Ronco T, Kappel LH, Aragao MF, et al. Insight into the antistaphylococcal activity of JBC 1847 at subinhibitory
concentration. Front Microbiol. 2022;12:786173.
48. Belley A, NeeshamGrenon E, McKay G, et al. Oritavancin kills stationaryphase and biofilm Staphylococcus aureus
cells in vitro.Antimicrob Agents Chemother. 2009;53(3):918925.
49. Lu CH, Shiau CW, Chang YC, et al. SC5005 dissipates the membrane potential to kill Staphylococcus aureus persisters
without detectable resistance. J Antimicrob Chemother. 2021;76(8):20492056.
50. Chen BC, Lin CX, Chen NP, Gao CX, Zhao YJ, Qian CD. Phenanthrene antibiotic targets bacterial membranes and
kills Staphylococcus aureus with a low propensity for resistance development. Front Microbiol. 2018;9:1593.
51. Crawford CL, Dalecki AG, Narmore WT, et al. Pyrazolopyrimidinones, a novel class of copperdependent bactericidal
antibiotics against multidrug resistant S. aureus.Metallomics. 2019;11(4):784798.
52. Gupta VK, Gaur R, Sharma A, et al. A novel bifunctional chalcone inhibits multidrug resistant Staphylococcus aureus
and potentiates the activity of fluoroquinolones. Bioorg Chem. 2019;83:214225.
53. Moon SH, Huang E. Novel linear lipopeptide paenipeptin Cbinds to lipopolysaccharides and lipoteichoic acid and
exerts bactericidal activity by the disruption of cytoplasmic membrane. BMC Microbiol. 2019;19(1):6.
54. Mumtaz S, Behera S, Mukhopadhyay K. Lipidated short analogue of αmelanocyte stimulating hormone exerts
bactericidal activity against the stationary phase of methicillinresistant Staphylococcus aureus and inhibits biofilm
formation. ACS Omega. 2020;5(44):2842528440.
55. Shi J, Chen C, Wang D, Tong Z, Wang Z, Liu Y. Amphipathic peptide antibiotics with potent activity against
multidrugresistant pathogens. Pharmaceutics. 2021;13(4):438.
56. Oyamada Y, Ito H, FujimotoNakamura M, et al. Anucleate cell blue assay: a useful tool for identifying novel type II
topoisomerase inhibitors. Antimicrob Agents Chemother. 2006;50(1):348350.
57. Trachtenberg MC, Packey DJ, Sweeney T. In vivo functioning of the Na
+
,K
+
activated ATPase. Curr Top Cell Regul.
1981;19:159217.
58. Makowski M, Felício MR, Fensterseifer ICM, Franco OL, Santos NC, Gonçalves S. EcDBS1R4, an antimicrobial
peptide effective against Escherichia coli with in vitro fusogenic ability. Int J Mol Sci. 2020;21(23):9104.
1086
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
59. Maisetta G, Vitali A, Scorciapino MA, et al. pHdependent disruption of Escherichia coli ATCC 25922 and model
membranes by the human antimicrobial peptides hepcidin 20 and 25. FEBS J. 2013;280(12):28422854.
60. Yang J, Yang H. Antibacterial activity of Bifidobacterium breve against Clostridioides difficile.Front Cell Infect Microbiol.
2019;9:288.
61. Sannasiddappa TH, Lund PA, Clarke SR. In vitro antibacterial activity of unconjugated and conjugated bile salts on
Staphylococcus aureus.Front Microbiol. 2017;8:1581.
62. Bruno ME, Kaiser A, Montville TJ. Depletion of proton motive force by nisin in Listeria monocytogenes cells. Appl
Environ Microbiol. 1992;58(7):22552259.
63. Tharmalingam N, Port J, Castillo D, Mylonakis E. Repurposing the anthelmintic drug niclosamide to combat
Helicobacter pylori.Sci Rep. 2018;8(1):3701.
64. Wu SC, Han F, Song MR, et al. Natural flavones from Morus alba against methicillinresistant Staphylococcus
aureus via targeting the proton motive force and membrane permeability. J Agricult Food Chem. 2019;67(36):
1022210234.
65. Begley M, Gahan CGM, Hill C. The interaction between bacteria and bile. FEMS Microbiol Rev. 2005;29(4):625651.
66. Webster CM, Woody AM, Fusseini S, Holmes LG, Robinson GK, Shepherd M. Proton motive force underpins
respirationmediated potentiation of aminoglycoside lethality in pathogenic Escherichia coli.Arch Microbiol.
2022;204(1):120.
67. Kappagoda S, Singh U, Blackburn BG. Antiparasitic therapy. Mayo Clin Proc. 2011;86(6):561583.
68. Chen W, Mook RA Jr., Premont RT, Wang J. Niclosamide: beyond an antihelminthic drug. Cell Signal. 2018;41:8996.
69. Silver LL. Challenges of antibacterial discovery. Clin Microbiol Rev. 2011;24(1):71109.
70. Hurdle JG, O'Neill AJ, Chopra I, Lee RE. Targeting bacterial membrane function: an underexploited mechanism for
treating persistent infections. Nat Rev Microbiol. 2011;9(1):6275.
71. Kim W, Zou G, Pan W, et al. The neutrally charged diarylurea compound PQ401 kills antibioticresistant and
antibiotictolerant Staphylococcus aureus.mBio. 2020;11(3):e0114020.
72. Kubo S, Niina T, Takada S. Molecular dynamics simulation of protontransfer coupled rotations in ATP synthase Fo
motor. Sci Rep. 2020;10(1):8225.
73. Zhao X, Kuipers OP. BrevicidineB, a new member of the brevicidine family, displays an extended target specificity.
Front Microbiol. 2021;12:693117.
74. Munita JM, Arias CA. Mechanisms of antibiotic resistance. Microbiol Spectr. 2016;4(2):VMBF0016201.
75. Crabbé A, Ostyn L, Staelens S, et al. Host metabolites stimulate the bacterial proton motive force to enhance the
activity of aminoglycoside antibiotics. PLoS Pathog. 2019;15(4):e1007697.
76. Liu Y, Tong Z, Shi J, Jia Y, Deng T, Wang Z. Reversion of antibiotic resistance in multidrugresistant pathogens using
nonantibiotic pharmaceutical benzydamine. Commun Biol. 2021;4(1):1328.
77. Ye J, Lin X, Cheng Z, et al. Identification and efficacy of glycine, serine and threonine metabolism in potentiating
kanamycinmediated killing of Edwardsiella piscicida.J Proteomics. 2018;183:3444.
78. Su Y, Peng B, Han Y, Li H, Peng X. Fructose restores susceptibility of multidrugresistant Edwardsiella tarda to
kanamycin. J Proteome Res. 2015;14(3):16121620.
79. Deng W, Fu T, Zhang Z, et al. Llysine potentiates aminoglycosides against Acinetobacter baumannii via regulation of
proton motive force and antibiotics uptake. Emerg Microbes Infect. 2020;9(1):639650.
80. Sun L, Sun L, Li X, et al. A novel tigecycline adjuvant ML7 reverses the susceptibility of tigecyclineresistant
Klebsiella pneumoniae.Front Cell Infect Microbiol. 2022;11:809542.
81. Magnowska Z, Jana B, Brochmann RP, et al. Carprofeninduced depletion of proton motive force reverses TetK
mediated doxycycline resistance in methicillinresistant Staphylococcus pseudintermedius.Sci Rep. 2019;9(1):17834.
82. Liu Y, Jia Y, Yang K, et al. Metformin restores tetracyclines susceptibility against multidrug resistant bacteria. Adv Sci.
2020;7(12):1902227.
83. Gupta P, Patel DK, Gupta VK, Pal A, Tandon S, Darokar MP. Citral, a monoterpenoid aldehyde interacts
synergistically with norfloxacin against methicillin resistant Staphylococcus aureus.Phytomedicine. 2017;34:8596.
84. Liu Y, Jia Y, Yang K, et al. Melatonin overcomes MCRmediated colistin resistance in Gramnegative pathogens.
Theranostics. 2020;10(23):1069710711.
85. Bhargava P, Collins JJ. Boosting bacterial metabolism to combat antibiotic resistance. Cell Metab. 2015;21(2):
154155.
86. Peng B, Li H, Peng XX. Functional metabolomics: from biomarker discovery to metabolome reprogramming. Protein
Cell. 2015;6(9):628637.
87. Yong Y, Zhou Y, Liu K, Liu G, Wu L, Fang B. Exogenous citrulline and glutamine contribute to reverse the resistance
of salmonella to apramycin. Front Microbiol. 2021;12:759170.
88. Ma Y, Guo C, Li H, Peng X. Low abundance of respiratory nitrate reductase is essential for Escherichia coli in
resistance to aminoglycoside and cephalosporin. J Proteomics. 2013;87:7888.
YANG ET AL.
|
1087
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
89. Maffei E, Fino C, Harms A. Antibiotic tolerance and persistence studied throughout bacterial growth phases.
Methods Mol Biol. 2021;2357:2340.
90. Van den Bergh B, Michiels JE, Wenseleers T, et al. Frequency of antibiotic application drives rapid evolutionary
adaptation of Escherichia coli persistence. Nat Microbiol. 2016;1:16020.
91. Mechler L, Herbig A, Paprotka K, Fraunholz M, Nieselt K, Bertram R. A novel point mutation promotes growth
phasedependent daptomycin tolerance in Staphylococcus aureus.Antimicrob Agents Chemother. 2015;59(9):
53665376.
92. LevinReisman I, Ronin I, Gefen O, Braniss I, Shoresh N, Balaban NQ. Antibiotic tolerance facilitates the evolution of
resistance. Science. 2017;355(6327):826830.
93. Wang M, Chan EWC, Wan Y, Wong MH, Chen S. Active maintenance of proton motive force mediates starvation
induced bacterial antibiotic tolerance in Escherichia coli.Commun Biol. 2021;4(1):1068.
94. Ciofu O, TolkerNielsen T. Tolerance and resistance of Pseudomonas aeruginosa biofilms to antimicrobial agentshow
P. aeruginosa can escape antibiotics. Front Microbiol. 2019;10:913.
95. Becker B, Cooper MA. Aminoglycoside antibiotics in the 21st century. ACS Chem Biol. 2013;8(1):105115.
96. Khan F, Pham DTN, Kim YM. Alternative strategies for the application of aminoglycoside antibiotics against the
biofilmforming human pathogenic bacteria. Appl Microbiol Biotechnol. 2020;104(5):19551976.
97. Kotra LP, Haddad J, Mobashery S. Aminoglycosides: perspectives on mechanisms of action and resistance and
strategies to counter resistance. Antimicrob Agents Chemother. 2000;44(12):32493256.
98. Walter F, Vicens Q, Westhof E. AminoglycosideRNA interactions. Curr Opin Chem Biol. 1999;3(6):694704.
99. Barza M, Scheife RT. Drug therapy reviews: antimicrobial spectrum, pharmacology and therapeutic use of antibiotics,
part 4: aminoglycosides. Am J Health Syst Pharm. 1977;34(7):723737.
100. Eliopoulos GM, Drusano GL, Ambrose PG, et al. Back to the future: using aminoglycosides again and how to dose
them optimally. Clin Infect Dis. 2007;45(6):753760.
101. Chopra I, Roberts M. Tetracycline antibiotics: mode of action, applications, molecular biology, and epidemiology of
bacterial resistance. Microbiol Mol Biol Rev. 2001;65(2):232260.
102. Keßler DN, Fokuhl VK, Petri MS, Spielmeyer A. Abiotic transformation products of tetracycline and chlortetracycline
in salt solutions and manure. Chemosphere. 2019;224:487493.
103. Nguyen F, Starosta AL, Arenz S, Sohmen D, Dönhöfer A, Wilson DN. Tetracycline antibiotics and resistance
mechanisms. Biol Chem. 2014;395(5):559575.
104. Seifert H, Blondeau J, Dowzicky MJ. In vitro activity of tigecycline and comparators (20142016) among key WHO
priority pathogensand longitudinal assessment (20042016) of antimicrobial resistance: a report from the T.E.S.T.
study. Int J Antimicro Ag. 2018;52(4):474484.
105. Gerson S, Nowak J, Zander E, et al. Diversity of mutations in regulatory genes of resistancenodulationcell division
efflux pumps in association with tigecycline resistance in Acinetobacter baumannii.J Antimicrob Chemother.
2018;73(6):15011508.
106. He F, Shi Q, Fu Y, Xu J, Yu Y, Du X. Tigecycline resistance caused by rpsJ evolution in a 59yearold male patient
infected with KPCproducing Klebsiella pneumoniae during tigecycline treatment. Infect Genet Evol. 2018;66:
188191.
107. Sun J, Chen C, Cui CY, et al. Plasmidencoded tet(X) genes that confer highlevel tigecycline resistance in Escherichia
coli.Nat Microbiol. 2019;4(9):14571464.
108. Tong Z, Xu T, Deng T, Shi J, Wang Z, Liu Y. Benzydamine reverses TMexCDTOprJmediated highlevel tigecycline
resistance in Gramnegative bacteria. Pharmaceuticals. 2021;14(9):907.
109. Lv L, Wan M, Wang C, et al. Emergence of a plasmidencoded resistancenodulationdivision efflux pump conferring
resistance to multiple drugs, including tigecycline, in Klebsiella pneumoniae.mBio. 2020;11(2):e0293019.
110. Redgrave LS, Sutton SB, Webber MA, Piddock LJV. Fluoroquinolone resistance: mechanisms, impact on bacteria, and
role in evolutionary success. Trends Microbiol. 2014;22(8):438445.
111. Azimi A, Rezaei F, Yaseri M, Jafari S, Rahbar M, Douraghi M. Emergence of fluoroquinolone resistance and possible
mechanisms in clinical isolates of Stenotrophomonas maltophilia from Iran. Sci Rep. 2021;11(1):9582.
112. Ghosh AS, Ahamed J, Chauhan KK, Kundu M. Involvement of an efflux system in highlevel fluoroquinolone
resistance of Shigella dysenteriae.Biochem Biophys Res Commun. 1998;242(1):5456.
113. Panta PR, Kumar S, Stafford CF, et al. A DedA family membrane protein is required for Burkholderia thailandensis
colistin resistance. Front Microbiol. 2019;10:2532.
114. Panta PR, Doerrler WT. A link between pH homeostasis and colistin resistance in bacteria. Sci Rep. 2021;
11(1):13230.
115. Capobianco JO, Goldman RC. Erythromycin and azithromycin transport into Haemophilus influenzae ATCC 19418
under conditions of depressed proton motive force (delta mu H). Antimicrob Agents Chemother. 1990;34(9):
17871791.
1088
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
116. Papkou A, Hedge J, Kapel N, Young B, MacLean RC. Efflux pump activity potentiates the evolution of antibiotic
resistance across S. aureus isolates. Nat Commun. 2020;11(1):3970.
117. Wang Y, Lu J, Mao L, et al. Antiepileptic drug carbamazepine promotes horizontal transfer of plasmidborne multi
antibiotic resistance genes within and across bacterial genera. ISME J. 2019;13(2):509522.
118. Zhang Y, Gu AZ, Cen T, Li X, Li D, Chen J. Petrol and diesel exhaust particles accelerate the horizontal transfer of
plasmidmediated antimicrobial resistance genes. Environ Int. 2018;114:280287.
119. Liu Y, Tong Z, Shi J, Jia Y, Yang K, Wang Z. Correlation between exogenous compounds and the horizontal transfer
of plasmidborne antibiotic resistance genes. Microorganisms. 2020;8(8):1211.
120. Lerminiaux NA, Cameron ADS. Horizontal transfer of antibiotic resistance genes in clinical environments. Can
J Microbiol. 2019;65(1):3444.
121. Li X, Wen C, Liu C, et al. Herbicide promotes the conjugative transfer of multiresistance genes by facilitating cellular
contact and plasmid transfer. J Environ Sci. 2022;115:363373.
122. Liao J, Huang H, Chen Y. CO
2
promotes the conjugative transfer of multiresistance genes by facilitating cellular
contact and plasmid transfer. Environ Int. 2019;129:333342.
123. Jia Y, Yang B, Shi J, Fang D, Wang Z, Liu Y. Melatonin prevents conjugative transfer of plasmidmediated antibiotic
resistance genes by disrupting proton motive force. Pharmacol Res. 2022;175:105978.
124. Johnston C, Martin B, Fichant G, Polard P, Claverys JP. Bacterial transformation: distribution, shared mechanisms
and divergent control. Nat Rev Microbiol. 2014;12(3):181196.
125. O'Neill AJ, Chopra I. Preclinical evaluation of novel antibacterial agents by microbiological and molecular techniques.
Expert Opin Invest Drugs. 2004;13(8):10451063.
126. Gentry DR, Wilding I, Johnson JM, et al. A rapid microtiter plate assay for measuring the effect of compounds on
Staphylococcus aureus membrane potential. J Microbiol Meth. 2010;83(2):254256.
127. Zhanel GG, Calic D, Schweizer F, et al. New lipoglycopeptides: a comparative review of dalbavancin, oritavancin and
telavancin. Drugs. 2010;70(7):859886.
128. Boix V, Fedorak RN, Mullane KM, et al. Primary outcomes from a phase 3, randomized, doubleblind, active
controlled trial of surotomycin in subjects with Clostridium difficile infection. Open Forum Infect Dis.
2017;4(1):ofw275.
129. Lee CH, Patino H, Stevens C, et al. Surotomycin versus vancomycin for Clostridium difficile infection: phase 2,
randomized, controlled, doubleblind, noninferiority, multicentre trial. J Antimicrob Chemother. 2016;71(10):
29642971.
130. Wang Z, Koirala B, Hernandez Y, et al. A naturally inspired antibiotic to target multidrugresistant pathogens. Nature.
2022;601(7894):606611.
131. Roberts KD, Zhu Y, Azad MAK, et al. A synthetic lipopeptide targeting toppriority multidrugresistant Gram
negative pathogens. Nat Commun. 2022;13(1):1625.
132. Velkov T, GallardoGodoy A, Swarbrick JD, et al. Structure, function, and biosynthetic origin of octapeptin antibiotics
active against extensively drugresistant Gramnegative bacteria. Cell Chem Biol. 2018;25(4):380391.
AUTHOR BIOGRAPHIES
Bingqing Yang graduated in 2020 as a Bachelor in veterinary medicine at Yangzhou University. Currently, she is
a postgraduate under the supervision of Prof. Yuan Liu at Yangzhou University. She is investigating the
inhibitors of horizontal transfer of antibiotic resistance genes and their mechanisms of action.
Ziwen Tong graduated in 2019 as a Bachelor in veterinary medicine at Xinjiang Agricultural University.
Currently, she is a postgraduate of Prof. Yuan Liu at Yangzhou University. She is investigating the combined use
of antibiotic adjuvants and existing antibiotics in the treatment of drugresistant infections.
Jingru Shi graduated in 2019 with a Bachelor in veterinary pharmacology at Henan Institute of Science and
Technology. Currently, she is a postgraduate of Prof. Yuan Liu at Yangzhou University. She is investigating the
synergistic basis of antimicrobial peptides in combination with antibiotics.
Zhiqiang Wang is a Professor of Veterinary Pharmacology and dean of the College of Veterinary Medicine at
Yangzhou University. He received his PhD degree in veterinary pharmacology in 2000 from South China
YANG ET AL.
|
1089
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Agricultural University. His research focuses on the transmission mechanisms and control strategies of drug
resistance of clinically important pathogens. To date, he has published more than 100 articles in peerreviewed
journals.
Yuan Liu is a Professor of Veterinary Pharmacology at the College of Veterinary Medicine at Yangzhou
University. He obtained his Bachelor's degree in veterinary medicine in 2013 from Southwest University and
achieved PhD degree in veterinary pharmacology in 2018 from China Agricultural University. His research
focuses on the identification of novel strategies to combat multidrugresistant (MDR) pathogens both in
preclinical studies and in the clinical setting, including the discovery of novel antimicrobial agents and antibiotic
adjuvants. To date, he has published more than 80 origin articles in this area, including Nature Microbiology,
Advanced Science, Angewandte Chemie International Edition, and Natural Product Reports.
How to cite this article: Yang B, Tong Z, Shi J, Wang Z, Liu Y. Bacterial proton motive force as an
unprecedented target to control antimicrobial resistance. Med Res Rev. 2023;43:10681090.
doi:10.1002/med.21946
1090
|
YANG ET AL.
10981128, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/med.21946 by Yangzhou University, Wiley Online Library on [06/06/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
... Membrane rigidity is associated with bacterial membrane homeostasis, especially the maintenance of PMF, which typically includes electrical potential (ΔΨ) and transmem brane proton gradient (ΔpH), is capable of regulating important biological functions such as adenosine triphosphate (ATP) synthesis and active transport of molecules (25). Bacterial PMF can not only act as an antibacterial target but also modulate conjugative transfer by affecting flagellar motility and energy metabolism (25); thus, we measured the changes in bacterial PMF after treatment with sub-MICs of Ind. First, we employed a fluorescent probe BCECF-AM to explore the effect of Ind on bacterial ΔpH. ...
... Membrane rigidity is associated with bacterial membrane homeostasis, especially the maintenance of PMF, which typically includes electrical potential (ΔΨ) and transmem brane proton gradient (ΔpH), is capable of regulating important biological functions such as adenosine triphosphate (ATP) synthesis and active transport of molecules (25). Bacterial PMF can not only act as an antibacterial target but also modulate conjugative transfer by affecting flagellar motility and energy metabolism (25); thus, we measured the changes in bacterial PMF after treatment with sub-MICs of Ind. First, we employed a fluorescent probe BCECF-AM to explore the effect of Ind on bacterial ΔpH. However, no fluorescence intensity changes were found (Fig. 4A), suggesting that sub-MICs Ind did not alter intracellular H + concentration. ...
Article
Full-text available
Antibiotic resistance represents a significant challenge to public health and human safety. The primary driver behind the dissemination of antibiotic resistance is the horizontal transfer of plasmids. Current conjugative transfer assay is generally performed in a standardized manner, ignoring the effect of the host environment. Host defense peptides (HDPs) possess a wide range of biological targets and play an essential role in the innate immune system. Herein, we reveal that sub-minimum inhibitory concentrations of HDPs facilitate the conjugative transfer of RP4-7 plasmid in the Luria Broth medium, and this observation is reversed in the RPMI medium, designed to simulate the host environment. Out of these HDPs, indolicidin (Ind), a cationic tridecapeptide from bovine neutrophils, significantly inhibits the conjugation of multidrug resistance plasmids in a dose-dependent manner, including blaNDM- and tet(X4)-bearing plasmids. We demonstrate that the addition of Ind to RPMI medium as the incubation substrate downregulates the expression of conjugation-related genes. In addition, Ind weakens the tricarboxylic acid cycle, impedes the electron transport chain, and disrupts the proton motive force, consequently diminishing the synthesis of adenosine triphosphate and limiting the energy supply. Our findings highlight the importance of the host-like environments for the development of horizontal transfer inhibitors and demonstrate the potential of HDPs in preventing the spread of resistance plasmids.
... On the other hand, the metabolism of bacteria is relied on transmembrane proton motive force (TPMF), which can be disrupted via consuming proton by OH − in an alkaline microenvironment, resulting in the generation of oxidative stress in bacteria to elevate intracellular ROS level and thus bacterial death. [20][21][22] Conceivably, the combination of enhanced ROS yield and the creation of an alkaline microenvironment can improve the bactericidal efficacy of the sonosensitive coating under US irradiation to a great extent. ...
... It is well established that the physiological microenvironment within the fractured bone, encompassing pH values and the concentrations of active ions, plays a key role in governing both bacterial-killing [20][21][22] and osteogenesis. [23][24][25] Accordingly, we examined the pH variations of the cell culture medium immersing the arrays. ...
Article
Full-text available
To effectively combat infectious osteoporosis, Ti‐based implants with enhanced antibacterial and osseointegrative properties are urgently required. Herein, a one‐step method involving Mg thermal‐reduction is employed to modify a hydroxyapatite (HA) array on Ti, which comprises an inner TiO2 layer and an outer nanorod‐like HA layer. This process introduces oxygen vacancies (OVs) into the TiO2 layer and deposits a Mg─O‐contained amorphous nanolayer on each HA nanorod. The introduced OVs enhance reactive oxygen species (ROS) yield by the array during ultrasound treatment via narrowing TiO2 bandgap and improving H2O molecules absorption. The produced ROS, combined with a weak alkaline microenvironment created by the degraded Mg─O nanolayer, endows the array with potent bacterial‐killing and biofilm‐eradicating efficacies within a 5 min ultrasound treatment via the combination of proton‐consumption and cell envelop‐detriment effects. Moreover, due to the alkaline microenvironment and the released Mg²⁺, the array hinders osteoclastogenesis by activating the inflammation‐related FAK‐PI3K‐AKT signaling pathway in macrophages, as revealed by transcriptomic analysis, resulting in robust osseointegration in rat femoral shaft with concurrent bacterial‐infection and osteoporosis. This work paves a new way for simultaneously endowing a sonosensitive coating on Ti with sonodynamic treatment‐derived high antibacterial ability and alkaline microenvironment‐mediated strong osseointegration for infectious osteoporosis treatment.
... Bacteria must maintain a proton motive force (PMF) for performing vital biochemical processes [53,54]. It was therefore important to investigate the effect of the three compounds alone or in combination on the bacterial membrane potential. ...
Article
Full-text available
Dental caries is a global health problem that requires better prevention measures. One of the goals is to reduce the prevalence of the cariogenic Gram-positive bacterium Streptococcus mutans. We have recently shown that naturally occurring arachidonic acid (AA) has both anti-bacterial and anti-biofilm activities against this bacterium. An important question is how these activities are affected by other anti-bacterial compounds commonly used in mouthwashes. Here, we studied the combined treatment of AA with chlorhexidine (CHX), cetylpyridinium chloride (CPC), triclosan, and fluoride. Checkerboard microtiter assays were performed to determine the effects on bacterial growth and viability. Biofilms were quantified using the MTT metabolic assay, crystal violet (CV) staining, and live/dead staining with SYTO 9/propidium iodide (PI) visualized by spinning disk confocal microscopy (SDCM). The bacterial morphology and the topography of the biofilms were visualized by high-resolution scanning electron microscopy (HR-SEM). The effect of selected drug combinations on cell viability and membrane potential was investigated by flow cytometry using SYTO 9/PI staining and the potentiometric dye DiOC2(3), respectively. We found that CHX and CPC had an antagonistic effect on AA at certain concentrations, while an additive effect was observed with triclosan and fluoride. This prompted us to investigate the triple treatment of AA, triclosan, and fluoride, which was more effective than either compound alone or the double treatment. We observed an increase in the percentage of PI-positive bacteria, indicating increased bacterial cell death. Only AA caused significant membrane hyperpolarization, which was not significantly enhanced by either triclosan or fluoride. In conclusion, our data suggest that AA can be used together with triclosan and fluoride to improve the efficacy of oral health care.
... Consistent with our findings, a previous study screened and identified potent inhibitors of S. pneumoniae competence, which regulated the transformation machinery and prevented gene transfer by inhibiting bacterial PMF [48]. These evidences revealed that PMF could serve as a critical target for the identification of HGT inhibitors [49]. ...
Article
Full-text available
The dissemination of antimicrobial resistance (AMR) severely degrades the performance of antibiotics and constantly paralyzes the global health system. In particular, plasmid-mediated transfer of antibiotic resistance genes (ARGs) across bacteria is recognized as the primary driver. Therefore, antiplasmid transfer approaches are urgently warranted to resolve this intractable problem. Herein, we demonstrated the potential of azidothymidine (AZT), an FDA-approved anti-HIV drug, as a broad-spectrum horizontal transfer inhibitor to effectively prevent the transmission of multiple ARGs, including mcr-1, blaNDM−5, and tet(X4), both in vitro and in vivo. It was also noteworthy that the inhibitory effect of AZT was proved to be valid within and across bacterial genera under different mating conditions. Mechanistic studies revealed that AZT dissipated bacterial proton motive force, which was indispensable for ATP synthesis and flagellar motility. In addition, AZT downregulated bacterial secretion systems involving general and type IV secretion systems (T4SS). Furthermore, the thymidine kinase, which is associated with DNA synthesis, turned out to be the potential target of AZT. Collectively, our work demonstrates the broad inhibitory effect of AZT in preventing ARGs transmission, opening new horizons for controlling AMR.
... Consistent with the effect of IBG on S. aureus, IBG increased fluorescence intensity in a concentration-dependent manner, . Bacterial PMF is an energy pathway located on the cell membrane of a bacterium and executes an important regulatory role in the synthesis of ATP, active transport of molecules, and rotation of bacterial flagellum (Yang et al., 2023). The PMF of bacteria binds sites and can be used to develop antibacterial agents and synergists (Hubbard et al., 2017;Stokes et al., 2020;Liu et al., 2021;Tong et al., 2021). ...
Article
Full-text available
Introduction: Riemerella anatipestifer (R. anatipestifer) is an important pathogen in waterfowl, leading to substantial economic losses. In recent years, there has been a notable escalation in the drug resistance rate of R. anatipestifer. Consequently, there is an imperative need to expedite the development of novel antibacterial medications to effectively manage the infection caused by R. anatipestifer. Methods: This study investigated the in vitro and in vivo antibacterial activities of a novel substituted benzene guanidine analog, namely, isopropoxy benzene guanidine (IBG), against R. anatipestifer by using the microdilution method, time-killing curve, and a pericarditis model. The possible mechanisms of these activities were explored. Results and Discussion: The minimal inhibitory concentration (MIC) range of IBG for R. anatipestifer was 0.5–2 μg/mL. Time-killing curves showed a concentration-dependent antibacterial effect. IBG alone or in combination with gentamicin significantly reduced the bacterial load of R. anatipestifer in the pericarditis model. Serial-passage mutagenicity assays showed a low probability for developing IBG resistance. Mechanistic studies suggested that IBG induced membrane damage by binding to phosphatidylglycerol and cardiolipin, leading to an imbalance in membrane potential and the transmembrane proton gradient, as well as the decreased of intracellular adenosine triphosphate. In summary, IBG is a potential antibacterial for controlling R. anatipestifer infections.
Article
Full-text available
The horizontal transfer of plasmids has been recognized as one of the key drivers for the worldwide spread of antimicrobial resistance (AMR) across bacterial pathogens. However, knowledge remain limited about the contribution made by environmental stress on the evolution of bacterial AMR by modulating horizontal acquisition of AMR plasmids and other mobile genetic elements. Here we combined experimental evolution, whole genome sequencing, reverse genetic engineering, and transcriptomics to examine if the evolution of chromosomal AMR to triclosan (TCS) disinfectant has correlated effects on modulating bacterial pathogen (Klebsiella pneumoniae) permissiveness to AMR plasmids and phage susceptibility. Herein, we show that TCS exposure increases the evolvability of K. pneumoniae to evolve TCS-resistant mutants (TRMs) by acquiring mutations and altered expression of several genes previously associated with TCS and antibiotic resistance. Notably, nsrR deletion increases conjugation permissiveness of K. pneumoniae to four AMR plasmids, and enhances susceptibility to various Klebsiella-specific phages through the downregulation of several bacterial defense systems and changes in membrane potential with altered reactive oxygen species response. Our findings suggest that unrestricted use of TCS disinfectant imposes a dual impact on bacterial antibiotic resistance by augmenting both chromosomally and horizontally acquired AMR mechanisms.
Article
Aims This study aimed to develop an editable structural scaffold for improving drug development, including pharmacokinetics and pharmacodynamics of antibiotics by using synthetic compounds derived from a (hetero)aryl-quinoline hybrid scaffold. Methods and results In this study, 18 CF3-substituted (hetero)aryl-quinoline hybrid molecules were examined for their potential antibacterial activity against Staphylococcus aureus by determining minimal inhibitory concentrations. These 18 synthetic compounds represent modifications to key regions of the quinoline N-oxide scaffold, enabling us to conduct a structure-activity relationship analysis for antibacterial potency. Among the compounds, 3 m exhibited potency against with both methicillin resistant S. aureus strains, as well as other Gram-positive bacteria, including Enterococcus faecalis and Bacillus subtilis. We demonstrated that 3 m disrupted the bacterial proton motive force (PMF) through monitoring the PMF and conducting the molecular dynamics simulations. Furthermore, we show that this mechanism of action, disrupting PMF, is challenging for S. aureus to overcome. We also validated this PMF inhibition mechanism of 3 m in an Acinetobacter baumannii strain with weaken lipopolysaccharides. Additionally, in Gram-negative bacteria, we demonstrated that 3 m exhibited a synergistic effect with colistin that disrupts the outer membrane of Gram-negative bacteria. Conclusions Our approach to developing editable synthetic novel antibacterials underscores the utility of CF3-substituted (hetero)aryl-quinoline scaffold for designing compounds targeting the bacterial proton motive force, and for further drug development, including pharmacokinetics and pharmacodynamics.
Article
Full-text available
Methicillin-resistant Staphylococcus aureus (MRSA) strains are tolerant of conventional antibiotics, making them extremely dangerous. Previous studies have shown the effectiveness of proton motive force (PMF) inhibitors at killing bacterial cells; however, whether these agents can launch a new treatment strategy to eliminate antibiotic-tolerant cells mandates further investigation. Here, using known PMF inhibitors and two different MRSA isolates, we showed that the bactericidal potency of PMF inhibitors seemed to correlate with their ability to disrupt PMF and permeabilize cell membranes. By screening a small chemical library to verify this correlation, we identified a subset of chemicals (including nordihydroguaiaretic acid, gossypol, trifluoperazine, and amitriptyline) that strongly disrupted PMF in MRSA cells by dissipating either the transmembrane electric potential (ΔΨ) or the proton gradient (ΔpH). These drugs robustly permeabilized cell membranes and reduced MRSA cell levels below the limit of detection. Overall, our study further highlights the importance of cellular PMF as a target for designing new bactericidal therapeutics for pathogens. IMPORTANCE Methicillin-resistant Staphylococcus aureus (MRSA) emerged as a major hypervirulent pathogen that causes severe health care-acquired infections. These pathogens can be multidrug-tolerant cells, which can facilitate the recurrence of chronic infections and the emergence of diverse antibiotic-resistant mutants. In this study, we aimed to investigate whether proton motive force (PMF) inhibitors can launch a new treatment strategy to eliminate MRSA cells. Our in-depth analysis showed that PMF inhibitors that strongly dissipate either the transmembrane electric potential or the proton gradient can robustly permeabilize cell membranes and reduce MRSA cell levels below the limit of detection.
Article
Full-text available
The emergence of multidrug-resistant (MDR) Gram-negative pathogens is an urgent global medical challenge. The old polymyxin lipopeptide antibiotics (polymyxin B and colistin) are often the only therapeutic option due to resistance to all other classes of antibiotics and the lean antibiotic drug development pipeline. However, polymyxin B and colistin suffer from major issues in safety (dose-limiting nephrotoxicity, acute toxicity), pharmacokinetics (poor exposure in the lungs) and efficacy (negligible activity against pulmonary infections) that have severely limited their clinical utility. Here we employ chemical biology to systematically optimize multiple non-conserved positions in the polymyxin scaffold, and successfully disconnect the therapeutic efficacy from the toxicity to develop a new synthetic lipopeptide, structurally and pharmacologically distinct from polymyxin B and colistin. This resulted in the clinical candidate F365 (QPX9003) with superior safety and efficacy against lung infections caused by top-priority MDR pathogens Pseudomonas aeruginosa, Acinetobacter baumannii and Klebsiella pneumoniae.
Article
Full-text available
Bacterial motility provides the ability for bacterial dissemination and surface exploration, apart from a choice between surface colonisation and further motion. In this study, we characterised the movement trajectories of pathogenic and probiotic Escherichia coli strains (ATCC43890 and M17, respectively) at the landing stage (i.e., leaving the bulk and approaching the surface) and its correlation with adhesion patterns and efficiency. A poorly motile strain JM109 was used as a control. Using specially designed and manufactured microfluidic chambers, we found that the motion behaviour near surfaces drastically varied between the strains, correlating with adhesion patterns. We consider two bacterial strategies for effective surface colonisation: horizontal and vertical, based on the obtained results. The horizontal strategy demonstrated by the M17 strain is characterised by collective directed movements within the horizontal layer during a relatively long period and non-uniform adhesion patterns, suggesting co-dependence of bacteria in the course of adhesion. The vertical strategy demonstrated by the pathogenic ATCC43890 strain implies the individual movement of bacteria mainly in the vertical direction, a faster transition from bulk to near-surface swimming, and independent bacterial behaviour during adhesion, providing a uniform distribution over the surface.
Article
Full-text available
Multidrug-resistant pathogens constitute a serious global issue and, therefore, novel antimicrobials with new modes of action are urgently needed. Here, we investigated the effect of a phenothiazine derivative (JBC 1847) with high antimicrobial activity on Staphylococcus aureus , using a wide range of in vitro assays, flow cytometry, and RNA transcriptomics. The flow cytometry results showed that JBC 1847 rapidly caused depolarization of the cell membrane, while the macromolecule synthesis inhibition assay showed that the synthesis rates of DNA, RNA, cell wall, and proteins, respectively, were strongly decreased. Transcriptome analysis of S. aureus exposed to sub-inhibitory concentrations of JBC 1847 identified a total of 78 downregulated genes, whereas not a single gene was found to be significantly upregulated. Most importantly, there was downregulation of genes involved in adenosintrifosfat (ATP)-dependent pathways, including histidine biosynthesis, which is likely to correlate with the observed lower level of intracellular ATP in JBC 1847–treated cells. Furthermore, we showed that JBC 1847 is bactericidal against both exponentially growing cells and cells in a stationary growth phase. In conclusion, our results showed that the antimicrobial properties of JBC 1847 were primarily caused by depolarization of the cell membrane resulting in dissipation of the proton motive force (PMF), whereby many essential bacterial processes are affected. JBC 1847 resulted in lowered intracellular levels of ATP followed by decreased macromolecule synthesis rate and downregulation of genes essential for the amino acid metabolism in S. aureus . Bacterial compensatory mechanisms for this proposed multi-target activity of JBC 1847 seem to be limited based on the observed very low frequency of resistance toward the compound.
Article
Full-text available
The increasing incidence of tigecycline resistance undoubtedly constitutes a serious threat to global public health. The combination therapies had become the indispensable strategy against this threat. Herein, 11 clinical tigecycline-resistant Klebsiella pneumoniae which mainly has mutations in ramR, acrR, or macB were collected for tigecycline adjuvant screening. Interestingly, ML-7 hydrochloride (ML-7) dramatically potentiated tigecycline activity. We further picked up five analogs of ML-7 and evaluated their synergistic activities with tigecycline by using checkerboard assay. The results revealed that ML-7 showed certain synergy with tigecycline, while other analogs exerted attenuated synergistic effects among tigecycline-resistant isolates. Thus, ML-7 was selected for further investigation. The results from growth curves showed that ML-7 combined with tigecycline could completely inhibit the growth of bacteria, and the time-kill analysis revealed that the combination exhibited synergistic bactericidal activities for tigecycline-resistant isolates during 24 h. The ethidium bromide (EtBr) efflux assay demonstrated that ML-7 could inhibit the functions of efflux pump. Besides, ML-7 disrupted the proton motive force (PMF) via increasing ΔpH, which in turn lead to the inhibition of the functions of efflux pump, reduction of intracellular ATP levels, as well as accumulation of ROS. All of which promoted the death of bacteria. And further transcriptomic analysis revealed that genes related to the mechanism of ML-7 mainly enriched in ABC transporters. Taken together, these results revealed the potential of ML-7 as a novel tigecycline adjuvant to circumvent tigecycline-resistant Klebsiella pneumoniae.
Article
Full-text available
It is well known that loss of aerobic respiration in Gram-negative bacteria can diminish the efficacy of a variety of bactericidal antibiotics, which has lead to subsequent demonstrations that the formation of reactive oxygen species (ROS) and the proton motive force (PMF) can both play a role in antibiotic toxicity. The susceptibility of Gram-negative bacteria to aminoglycoside antibiotics, particularly gentamicin, has previously been linked to both the production of ROS and the rate of antibiotic uptake that is mediated by the PMF, although the relative contributions of ROS and PMF to aminoglycoside toxicity has remained poorly understood. Herein, gentamicin was shown to elicit a very modest increase in ROS levels in an aerobically grown Escherichia coli clinical isolate. The well-characterised uncoupler 2,4-dinitrophenol (DNP) was used to disrupt the PMF, which resulted in a significant decrease in gentamicin lethality towards E. coli . DNP did not significantly alter respiratory oxygen consumption, supporting the hypothesis that this uncoupler does not increase ROS production via elevated respiratory oxidase activity. These observations support the hypothesis that maintenance of PMF rather than induction of ROS production underpins the mechanism for how the respiratory chain potentiates the toxicity of aminoglycosides. This was further supported by the demonstration that the uncoupler DNP elicits a dramatic decrease in gentamicin lethality under anaerobic conditions. Together, these data strongly suggest that maintenance of the PMF is the dominant mechanism for the respiratory chain in potentiating the toxic effects of aminoglycosides.
Article
Full-text available
Gram-negative bacteria are responsible for an increasing number of deaths caused by antibiotic-resistant infections1,2. The bacterial natural product colistin is considered the last line of defence against a number of Gram-negative pathogens. The recent global spread of the plasmid-borne mobilized colistin-resistance gene mcr-1 (phosphoethanolamine transferase) threatens the usefulness of colistin³. Bacteria-derived antibiotics often appear in nature as collections of similar structures that are encoded by evolutionarily related biosynthetic gene clusters. This structural diversity is, at least in part, expected to be a response to the development of natural resistance, which often mechanistically mimics clinical resistance. Here we propose that a solution to mcr-1-mediated resistance might have evolved among naturally occurring colistin congeners. Bioinformatic analysis of sequenced bacterial genomes identified a biosynthetic gene cluster that was predicted to encode a structurally divergent colistin congener. Chemical synthesis of this structure produced macolacin, which is active against Gram-negative pathogens expressing mcr-1 and intrinsically resistant pathogens with chromosomally encoded phosphoethanolamine transferase genes. These Gram-negative bacteria include extensively drug-resistant Acinetobacter baumannii and intrinsically colistin-resistant Neisseria gonorrhoeae, which, owing to a lack of effective treatment options, are considered among the highest level threat pathogens⁴. In a mouse neutropenic infection model, a biphenyl analogue of macolacin proved to be effective against extensively drug-resistant A. baumannii with colistin-resistance, thus providing a naturally inspired and easily produced therapeutic lead for overcoming colistin-resistant pathogens.