Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

| |

Received: 4 October 2021    Revised: 28 December 2021    Accepted: 11 February 2022

DOI: 10.1111/jam.15492

REVIEW ARTICLE

Heavy metal-­induced selection and proliferation of


antibiotic resistance: A review

Prakriti Vats  | Ujjwal Jit Kaur  | Praveen Rishi

Department of Microbiology, Panjab Abstract


University, Chandigarh, India
Antibiotic resistance is recognized as a global threat to public health. The selection
Correspondence and evolution of antibiotic resistance in clinical pathogens were believed to be ma-
Prof. Praveen Rishi, Department of jorly driven by the imprudent use of antibiotics. However, concerns regarding the
Microbiology, BMS Block I, South
Campus, Panjab University, Sector 25,
same, through selection pressure by a multitude of other antimicrobial agents, such
Chandigarh, 160014, India. as heavy metals, are also growing. Heavy metal contamination co-­selects antibiotic
Email: rishipraveen@yahoo.com and metal resistance through numerous mechanisms, such as co-­resistance and
cross-­resistance. Here, we have reviewed the role of heavy metals as antimicrobial re-
sistance driving agents and the underlying concept and mechanisms of co-­selection,
while also highlighting the scarcity of studies explicitly inspecting the process of co-­
selection in clinical settings. Prospective strategies to manage heavy metal-­induced
antibiotic resistance have also been deliberated, underlining the need to find spe-
cific inhibitors so that alternate medicinal combinations can be added to the existing
therapeutic armamentarium.

KEYWORDS
antibiotics, antimicrobials, environment health, heavy metals, resistance

I N T RO DU CT ION wherein even minor infections will be life-­threatening


(WHO, 2014). Subsequently, to control the emergence and
The discovery of antibiotics and the exploitation of their propagation of antibiotic-­resistant strains, various mea-
ability to treat infectious diseases has entirely revolution- sures to promote the judicious use of antibiotics have been
ized the field of public health. However, the relentless use established. However, these measures have encountered
of antibiotics, along with the endless adaptive capabilities limited success and the phenomenon of antibiotic resis-
of micro-­organisms, formed the basis for the selective pro- tance among pathogens is still prevailing (Gillespie, 2001;
liferation and evolution of antibiotic-­resistant bacteria. Sundqvist et al., 2009). This indicates the presence of fac-
This has ultimately triggered the advent of ‘multidrug-­ tors, other than the antibiotics, that may govern the evo-
resistant bacteria (MDR)’(Aminov,  2010; Lehtinen lution, selection and dissemination of antibiotic-­resistant
et al., 2019). Thus, the medical community is encounter- strains (Baker-­Austin et al., 2006).
ing a challenge to treat microbial infections with the cur- Resistance in pathogens is mediated by both the in-
rently available array of antibiotics, as resistance due to trinsic (chromosomal) and extrinsic (extrachromosomal)
antibiotics is spreading among pathogenic bacteria at an factors. Extrachromosomal resistance is usually held re-
unprecedented rate (Aslam et al., 2018). sponsible for antimicrobial resistance (AMR) in environ-
Antibiotic treatment failure accounts for hundreds mental micro-­organisms. The evidence available suggests
of thousands of deaths each year. WHO has declared that such resistance determinants are transferred between
antibiotic resistance as a global health crisis that, if not the environment and the clinical disease-­causing bacte-
controlled urgently, can lead us into a postantibiotic era, ria. In this context, the role of the natural environment in

| 2022 Society for Applied Microbiology.


4058    © wileyonlinelibrary.com/journal/jam J Appl Microbiol. 2022;132:4058–4076.
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4059

the selection and evolution of antibiotic resistance came Consequently, this review focuses on the current knowl-
into focus. A variety of other substances, such as heavy edge regarding the indispensable role of heavy metals in
metals, occurring in the natural environment, may trig- the co-­selection and dissemination of antibiotic-­resistant
ger this process (Romero et al., 2017; Skurnik et al., 2010). bacteria. Maintenance of horizontally transferable ARG
Bacteria are continuously exposed to heavy metals pres- pools and the emergence of multidrug resistance (MDR) in
ent in the natural habitats due to the ungoverned release natural and clinical settings are also discussed along with
of industrial and agricultural effluents, thereby resulting potential molecular mechanisms behind the co-­selection
in the survival of the metal-­tolerant cells due to muta- process and the prospective strategies to disarm it.
tions ­(Al-­Musharafi,  2016; Masindi and Muedi,  2018).
Resistance to heavy metals in pathogens has been asso-
ciated with the acquisition of extrinsic resistance deter- ROLE OF ANTIMIC ROBIAL
minants. Antibiotic Resistance genes (ARGs) containing RESISTANCE DRIVING AGENT S :
Class 1 integrons and gene cassettes in microbes have FROM THE ENV IRONMENT TO
been reported in samples collected from sewage treatment C LINICS
plants, which indicates the co-­selection of metal and an-
tibiotic resistance-­associated genes (Zhang et al.,  2009; The estimated number of bacteria on earth is in the order
Kotlarska et al., 2015). Such co-­selection also raises serious of magnitude of 1030 (Whitman et al., 1998). Their omni-
concerns regarding ungoverned dumping of metal-­ridden presence, in such a generous amount, abets their interac-
industrial and agricultural effluents. Once these effluents tion with a vast chemical diversity in the environment,
are dumped in the open, they form waste reservoirs which which may be of abiotic origin or produced by other bac-
contribute towards the maintenance and transfer of ARGs teria, fungi, plants, or animals. Many of these chemical
in pathogens. compounds have an impact on the cellular processes of
The first few reports indicating the correlation between bacteria in their vicinity, by which, they become the driv-
the selection of multiple antibiotics and heavy metal resis- ers of natural selection and evolution of genetic diversity
tance were published as early as the 1960s (Moore, 1960; of these bacteria (Martinez, 2009; Wright, 2010). The re-
Richmond and John, 1964). Thereafter, a significant num- sulting impact of constant exposure of environmental
ber of such studies have been documented which suggest chemicals on the bacteria was the development of com-
the role of heavy metals in the co-­selection and proliferation plex mechanisms and novel proteins for sensing, respond-
of antibiotic-­resistant bacteria (Bhattacharya et al.  2000, ing to, and metabolizing small molecules. Some of these
Pathak & Gopal,  2005). Peltier et al.  (2010) reported en- proteins having modest antibiotic resistance functions or
hancement of antibiotic resistance in the microbial com- some affinity for antibiotics may evolve into a bona fide
munity through the development of cross-­resistance via resistance mechanism in response to antibiotic selective
co-­exposure to subtoxic levels of zinc and some unrelated pressure and thus, act as precursors to resistance pro-
antibiotics, such as ciprofloxacin, oxytetracycline and teins (Kaufmann et al., 2005; Yim et al., 2006; Fajardo and
tylosin, in laboratory-­scale wastewater treatment bio- Martínez, 2008).
reactors. The addition of copper to agricultural soil, in a The environmental organisms that produce chemical
field-­based study, was reported to indirectly select anti- substances or secondary metabolites, such as, antibiotics,
biotic resistance along with copper resistance by encour- have to protect themselves from the toxicity of these mol-
aging horizontal gene transfer (HGT) of mobile genetic ecules also, and hence, these become the hotspots for the
elements (MGEs) (Berg et al., 2010). Furthermore, certain evolution of resistance genes. Their neighbouring micro-­
reports indicate that subinhibitory concentrations of heavy organisms must also develop coping strategies to survive
metals in the environment enhance the frequency of anti- in a toxic environment. This is done either through the
biotic resistance by inducing mutagenesis in bacteria (Li evolution of resistance mechanisms or by the HGT of re-
et al., 2019; Li et al., 2021). Hence, several studies substan- sistance genes from the producer organism (Benveniste
tiate the claims under the co-­selection theory and establish and Davies, 1973; Wright, 2007).
a relation between heavy metals and antibiotic resistances. Genome sequencing of all bacteria, even with modest
Many of these studies direct that relatively low concentra- genomes, shows the presence of specific genes responsible
tions of these heavy metals in the polluted environment for metabolizing small chemical molecules of foreign or-
suffice to induce antibiotic resistance (Chen et al., 2015), igin for nutrition and defence (Fraser et al., 1995). These
which is a matter of grave concern for the medical commu- genes encode outer membrane transport and efflux pro-
nity considering that much higher levels of heavy metals teins, enzymes to metabolize specific molecules, and re-
from anthropogenic sources exist in both natural and clin- ceptor molecules. Microbes can incorporate any of these
ical environments (Stepanauskas et al., 2005). classes of genes in response to cytotoxic molecules such
|
4060       VATS et al.

as heavy metals or antibiotics. These chromosomal genes pressure, the MGEs rapidly became the vectors for car-
are the reservoir for resistance genes and comprise the rying resistance genes. The majority of the resistance
environmental resistome. Therefore, it is apparent that genes found in pathogenic bacteria infecting mammals
micro-­organisms that evolved under constant exposure are carried by the MGEs, such as plasmids, which are
to diverse chemical molecules such as antibiotics, metal acquired by HGT from the environmental bacteria. A
ions and other secondary metabolites become the source significant number of reports are available that suggest
of clinically relevant ARGs and such chemical molecules a higher prevalence of antibiotic resistance in commen-
become the drivers of AMR. sal bacteria harboured by wild animals dwelling close
The huge diversity of ARGs, fostered by the environ- to human habitats (OÖsterblad et al., 2001). A report by
mental resistome, was neglected to a large extent by the Skurnik et al.  (2006) demonstrated a clear correlation
scientific community for a long time because the epide- between high antibiotic resistance frequency in E. coli
miology of antibiotic resistance was limited to clinical set- sampled from farm animals with increased human expo-
tings. Ample studies emphasize the fact that the resistance sure. On the same lines, higher antibiotic resistance was
genes responsible for MDR can originate in nonpatho- also recovered from animals, such as gorillas, baboons
genic environmental microbes in nonclinical settings and rodents which lived close to human habitats and
(Rutgersson et al.,  2014; Xiong et al.,  2014). This is best livestock, whereas unexposed counterparts, which lived
illustrated by the example of ARGs coding for aminogly- away from human habitats showed practically no antibi-
coside and vancomycin whose working is almost similar otic resistance (Rolland et al., 1985; Gilliver et al., 1999;
in both environmental and clinical isolates. The environ- OÖsterblad et al., 2001; Rwego et al., 2008). These stud-
mental counterparts of these ARGs upon acquisition by ies imply that the enduring concentrations of antibiot-
human pathogens would eventually evolve into explicit ics present in such habitats select the ARGs and lead to
antibiotic resistance determinants under specific antibi- their spread and diversification among micro-­organisms
otic selection pressures posed by humans. living within the same geographical boundaries.
The prevalence of ARGs is narrowed down while pro- All in all, the presence of high concentrations of an-
ceeding towards clinics from environmental resistome as tibiotics serves as a selection pressure on the microbes
they adapt to selection pressures that are specific to clini- in the vicinity, aiding the maintenance and employment
cal settings. Since ARGs are highly prevalent in the envi- of resistance genes in the environmental bacteria. It
ronment, surveillance of natural environments should be should be noted that the environment is also a source
done regularly to monitor antibiotic resistance as it can of opportunistic pathogens which may already be re-
serve as a warning system to assess imminent antibiotic sistant or may acquire resistance genes via HGT from
resistance mechanisms (D’Costa et al., 2006) and also help human-­associated bacteria and subsequently cause in-
us understand the pattern of prevalence, distribution and fections in humans. During all these processes, a major
transmission of ARGs (Wright, 2010). concern for the clinical community arises when novel
resistance genes, originally absent in the resistance
genes repertoire of pathogens, are recruited from the
T H E E NV I RO N M E N TAL environmental resistome, thus posing a challenge to
RES I STO M E AN D H U M AN the existing therapies and severe consequences for the
PAT H O G E NS infected patients. Several studies support this by ev-
idencing the existence of identical resistance genes in
All the ARGs circulating in the environment among human pathogens and the environmental bacterial iso-
antibiotic producers and nonpathogenic environmental lates. For example a metagenomic study by Forsberg
bacteria constitute the environmental resistome. The et al.  (2012) reported several ARGs present in human
environmental resistome predates the therapeutic use pathogens, conferring resistance to antibiotics, such as
of antibiotics in humans. The widely distributed ARGs β-­lactams, sulphonamides, aminoglycosides, phenicol
must express in disease-­causing bacteria to pose a clini- and tetracyclines, which were similar to bacteria from
cal threat, establishing the point of intersection for the various U.S. soils. Reports with similar claims have also
environmental resistome and the resistome of patho- been published by Poirel et al.  (2004, 2005). These re-
genic organisms. Anthropogenic sources of antibiotics ports emphasize that the Gram-­negative bacteria from
have significantly increased in clinical settings as well as the environmental resistome is the reservoir for future
human habitats, thus compelling the human pathogens ARGs which will ultimately transfer to human patho-
to acquire antibiotic resistance elements from the envi- gens through HGT due to overlapped geographical
ronmental resistome. In the face of antibiotic selective niches and cause infections in humans.
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4061

H E AV Y M ETALS: T HE AG E N T S OF forest fires are more examples of natural processes that


CO - ­S E L ECT ION allow the mobilization of metals in the environment. All
these natural processes help in the cycling of metals in the
Microbes frequently encounter metals in their habitats. ecosystem and thus, ensure their constant background
This interaction can either be beneficial or harmful for levels in the environment (Mighall et al., 2002; Raab and
the microbe (Ehrlich, 1997). Based on utility, heavy met- Feldmann, 2003).
als may be classified as ‘essential micronutrients’ and In addition to the natural sources, three major anthro-
‘nonessential metals’. Chromium (Cr), calcium (Ca), zinc pogenic sources contribute to the heavy metal pollution
(Zn), copper (Cu), manganese (Mn), iron (Fe), potassium in the environment: Industries, agriculture and sewage
(K), magnesium (Mg), nickel (Ni), sodium (Na) and co- sludge. Lately, there has been an unprecedented increase
balt (Co) are the ‘essential’ metals, which are involved in in ungoverned industrial activities that have intensified
the fundamental metabolic processes of micro-­organisms. the problem of environmental pollution along with the
These metals play important roles in redox reactions in accumulation of toxic heavy metals in air, water and soil.
cells, as cofactors of enzymes, as stabilizers, and have a Industrial activities, such as mining of ores, metal smelt-
role in regulating cellular osmotic pressure. Whereas ing, activities of metallurgical industries, refining, the
other metals, for example gold (Au), silver (Ag), antimony leftover waste of mining and metallurgical processes, elec-
(Sb), cadmium (Cd), arsenic (As), lead (Pb), aluminium troplating, plastics manufacturing, fertilizer-­producing
(Al) and mercury (Hg) do not have any crucial function plants, thermal power plants and corrosion of metal
in biological processes and hence, are considered as ‘non- equipment in use, have led to an elevated concentration
essential metals’. Therefore, they are extremely toxic to of metals in different trophic levels of the ecosystem
micro-­organisms and are used as antimicrobials, having (Zouboulis et al., 2004; Oyetibo et al., 2010).
a broad spectrum of activity. Even essential heavy met- Agriculture and aquaculture are also major burdens
als, such as Cu, Zn and Ni, at high concentrations form to the environment in terms of metal pollution. Usage
nonspecific complexes within the cells and eventuate of antimicrobial compounds, pesticides, fertilizers and
toxicity. Toxicity heavily depends on the environmental sewage sludge, all of which contain mineral nutrients
conditions of the cell, for instance, the pH, redox poten- for plant and animal growth, acts as a major contributor
tial and concentration of organic matter, as these affect to the problem. Adding sewage sludge to soil serves as a
the valency and bioavailability of the metals (Nies, 1999; low-­cost disposal technique as well as a natural fertilizer
Seiler and Berendonk, 2012). The elevation in both intra to the crops. Heavy metals, such as Hg, Cu, As, Zn, Cr, Pb
and extra-­cellular levels of metals in bacterial cells can be and Cd, are present in a wide range of concentrations in
detrimental for the activity of enzymes in the cell, basic both animal feed supplements and plant manures with Zn
cellular functions, may damage the cellular membranes, and Cu being one of the most accumulated metals in ani-
and may even permanently damage the structure of mal feed as they are commonly used as growth enhancers
DNA. Maintaining heavy metal homeostasis is essential (Yazdankhah et al.,  2014). These metals have antimicro-
for the survival of the cell and hence, the concentration bial properties similar to antibiotics. Thus, they suppress
of heavy metals inside the cell is under strict regulation pathogens and fermentation processes in the gut which in
(Nies, 1999). Thus, metal toxicity poses a selection pres- turn promotes growth by preventing the loss of nutrients.
sure to the bacterial community. However, utilizing their Such high levels of Cu and Zn in the feed supplements are
infinite adaptive capabilities, bacteria have evolved sys- poorly absorbed by the animals and hence, most of them
tems for resistance against almost all toxic metals which are excreted in the faeces. From there, these metals may
helps them survive and selectively proliferate in a toxic, directly enter the soil environment or may be added later
heavy metal-­ridden environment (Rouch et al., 1995). to the soil through manure application. Once these nutri-
ents reach the soil, they may contaminate groundwater as
well through leaching. The addition of these nutrients to
H E AV Y M ETAL POLLU T ION : A soil has been reported to select for metal as well as indi-
WO RL DWI DE PROB LE M rectly for antibiotic resistance (Hu et al., 2017).
Contamination of marine environments and surface
Metals are naturally present in the earth’s crust and infil- water by metals is mainly accounted for by aquaculture
trate the biome through natural as well as certain anthro- practices and sewage. Fish farmers usually use antimicro-
pogenic activities. Natural processes, such as weathering bial metals, metal-­containing anti-­fouling agents, or fish
or erosion by wind and water, keep transmitting signifi- food enriched with micronutrients. Cu-­based paints are
cant amounts of natural metals into the ecosystem. The the most commonly used antifouling paints in the aqua-
aerosol formation above the sea, volcanic eruptions, and culture industry (Burridge et al., 2010). Cu alloys are also
|
4062       VATS et al.

used in the netting material (Flach et al.,  2017). Zinc is urgent introduction of efficacious strategies and policies
used as a supplement for salmon feed. Marine sediments is required to control it. In addition to this, heavy metals,
in the periphery of fish farms, thus, have high concentra- being nonbiodegradable, enter the food chain via contam-
tions of metals such as Cu and Zn. Five different drugs ap- inated soil and water and end up accumulating in different
proved by the FDA, which are sulfamerazine, florfenicol, trophic levels, thus, also posing a direct threat to human
oxytetracycline dihydrate, oxytetracycline hydrochloride, health and environmental biodiversity.
chorionic gonadotropin as well as sulfadimethoxine and
ormetoprim in combination, are routinely used in aqua-
culture (Seiler and Berendonk, 2012). Thus, prospects of CO - ­S ELECTION OF METAL AND
selection and co-­selection of antibiotic resistance in bacte- ANTIBIOTIC RESISTANC E
rial communities in aquaculture are very high as they are
repeatedly exposed to a combination of heavy metals and As opposed to antibiotics, metals have biologically ex-
antibiotics. isted since the formation of the earth and the great oxida-
Continuous release of industrial and agricultural tion event nearly 2.4 billion years ago (Fru et al.,  2016).
waste, having abundant heavy metal content, into sew- Accordingly, the genes associated with toxic metal re-
age contaminates sewage water as well. Sewage water sistance are also believed to be primordial (Jackson and
provides optimum conditions for the proliferation of Dugas, 2003; Boyd and Barkay, 2012). Even then, antibi-
antibiotic-­resistant bacteria. An abundance of ARGs and otic resistance was discovered first and not metal resist-
MGEs in sewage water and mining affected areas has also ance. Mercury resistance transposons, having a close
been reported in many studies (Pazda et al.,  2019; Zou resemblance to contemporary Tn21-­like transposons, but
et al., 2021). Hence, sewage water acts as a perfect hotspot lacking ARGs, have been isolated recently from ‘preanti-
for HGT of MGEs containing ARGs in the presence of biotic era’ bacteria (Essa et al., 2003; Kholodii et al., 2003).
metals, thereby promoting co-­selection of metal and an- Whole-­genome sequencing of ‘Preantibiotic era’ bacteria,
tibiotic resistance. Co-­selection can be triggered once the resuscitated from long-­term storage, has also corroborated
metals accumulate to a critical concentration. the presence of multiple metal resistance genes (MRGs)
In the case of clinical settings, routine cleaning of (Dunne et al., 2017). Therefore, it can be concluded that
high-­touch surfaces around patients does not get rid of metal resistance is a fairly pre-­existing feature of heavy
contaminating pathogens, causing major healthcare-­ metal exposed microbial communities, which might have
associated infections (HAIs) (Boyce,  2007). To mitigate somehow influenced or promoted the rapid modern-­day
this problem, such surfaces are covered with antimicro- evolution of AMR (Pal et al., 2017).
bial coatings containing metals such as Cu and Ag, or The co-­selection of metal and antibiotic resistance is
antimicrobial peptides (Page et al., 2009). However, such instituted on the theoretical similarity and overlapping
metal-­ridden surfaces provide the bacterial community between the resistance mechanism for both these an-
with a heavy metal selection pressure and hence, may timicrobial agents. The first account of acquired heavy
form suitable platforms for co-­selection of antibiotic and metal resistance in bacteria was given by Moore  (1960)
metal resistance (Pietsch et al., 2020). In addition to this, after isolating mercury-­resistant S. aureus from an epi-
hospital waste, such as damaged thermometers, barome- demic hospital infection. However, the genetic linkage of
ters, sphygmomanometers or other waste electrical equip- acquired mercury resistance in S. aureus, isolated from
ment and biomedical waste, is usually ridden with heavy wound infections with high penicillinase activity was
metals. Thus, hospitals and clinics also act as a significant described by Richmond and John (1964). Furthermore,
source of heavy metal pollution. Timoney et al. (1978) recovered Bacillus strains from sew-
The concentration of antibiotics in aquatic (Kolpin age sludge which showed combined resistance to mercury
et al., 2002), as well as soil environments, is generally and ampicillin frequently. These Bacillus strains suggest
low as they are continuously being degraded rapidly via a simultaneous selection of genes for both these resis-
photolytic and other pathways (Cardoza et al., 2005). But tances under a mercury selection pressure in the sludge.
depending on bioavailability and chemical forms, metals Decade-­old profound researches on S. aureus, E. coli, and
are highly prevalent in most environments and are ever-­ S. Typhimurium have also revealed a correlation of anti-
increasing (Stepanauskas et al., 2006; Berg et al.,  2010). biotic and metal resistance as the ARGs carrying plasmids
This implies that heavy metals, being highly recalcitrant, of these human pathogens also carried MRGs, indicating
can provide the bacterial community with a long-­lasting a co-­selection potential (Novick and Roth,  1968; Hedges
selection pressure (Stepanauskas et al.,  2006). Thus, and Baumberg, 1973; Hobman and Crossman, 2015).
heavy metal contamination of the environment has a Since metals are ubiquitous, posing an enduring selec-
far-­reaching impact on the dissemination of ARGs and tion pressure on microbes, a positive correlation between
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4063

the presence of metals and ARGs has been described in (Y. Zhang et al., 2018; S. Zhang et al., 2019; Lu et al., 2020;
several environmental contexts, for example in agricul- Wang et al.,  2020). This facilitates the transmission of
tural soils, mining-­affected areas, aquaculture systems, antibiotic resistance among environmental bacteria and
swine and swine farms, in dairy farms and the gut mi- subsequently, the pathogenic bacteria as well. All these
crobiota of various other animals (Resende et al.,  2012; studies connote that metal exposure can co-­select for an-
Holman and Chénier,  2015; Zhou et al.,  2016; Ding tibiotic resistance in clinically relevant bacterial isolates.
et al., 2019; Maurya et al., 2020; Zou et al., 2021). The use The primary benefit that the bacteria realize from co-­
of metals, such as Zn and Cd, as therapeutics in animal selection is the ability to sustain a multitude of antimicro-
feeds, instead of or as a supplement to antibiotics, was bial selection pressures which will help in the subsistence,
shown to co-­select and increase the prevalence of meth- proliferation and evolution of different bacterial species
icillin resistance in S. aureus. This was attributed to the in the environment. Co-­selection makes bacteria more fit
co-­located genes for metal resistance (czr) and methicil- for survival. Furthermore, in case such a bacterium, har-
lin resistance (mecA) on staphylococcal cassette chromo- bouring resistance against multiple antimicrobial agents,
some mec (SSCmec) (Cavaco et al.,  2010, 2011). Similar including antibiotics, enters the clinical settings, it would
co-­selection of zinc and methicillin-­resistant S. aureus be difficult to eradicate it. Such drug-­resistant bacteria
(MRSA) has been reported in the microbiota of pigs as have the potential to elicit hospital epidemics, critical
well (Hau et al., 2017; Slifierz et al., 2015). Environmental infections, sepsis, and the failure of antibiotic therapies.
isolates of E. coli frequently display multiple antibiotic re- Multidrug resistance is already prevalent in hospitals. In
sistance on exposure to metals, for example resistance to addition to this, exposure to multiple antibiotics, in an
β-­lactams on exposure to copper and zinc in pig manure attempt to treat the existing problem, can further aid in
(Hölzel et al., 2012), and increase in resistant isolates of developing resistance to other drugs/ antibiotics which
E. coli in the gut microbiota of piglets on supplementation are used clinically. Therefore, the comprehension of the
of zinc (Bednorz et al., 2013). The genes for streptomycin co-­selection process is imperative as it has the potential to
and mercury resistance are reported to be present on Tn21 sustain and promote ARGs even in the dearth of antibiot-
transposon in isolates of avian E. coli, thus indicating that ics, having a long-­term clinical and environmental impact.
both these genes have a potential of co-­transferability
which may lead to the selection of streptomycin resistance
on exposure to mercuric compounds (Bass et al., 1999). THE CONCEPT BEHIND THE
IncHI-­type plasmids, containing multiple resistance CO - ­S ELECTION OF RESISTANC E
genes, isolated from both humans and animals, and pres- GENES
ent in Gram-­negative bacteria of the Enterobacteriaceae
family, have been linked to the propagation of metal and The ability of the micro-­organisms to undergo minute
antibiotic resistance. One such plasmid of the IncHI-­2 changes that allow their survival in extreme conditions,
group, namely pMG101 (182  kb), conferring resistance such as strong antimicrobial selection pressures, eventu-
to ampicillin, streptomycin, sulphonamide, chloram- ally results in the evolution of resistance. These changes
phenicol, tetracycline, tellurium, mercury and silver was may either be chromosomal mutations or the acquisition
isolated from Salmonella enterica serovar Typhimurium of MGEs, which confer resistance to the microbial cell.
(Larkin Mchugh et al., 1975; Gupta et al., 1999). Although these changes allow the cell to survive, they be-
Various experimental as well as in situ studies, in- stow a metabolic burden on the cell, which makes it less
vestigating the co-­selection process in the environment, fit to survive in the absence of selection pressure. Thus,
have also been carried out by several researchers (Berg such changes in a bacterial cell that allow it to survive in
et al.,  2005; M. Zhang et al.,  2020; Silva et al.,  2021). conditions of stress, give rise to its ‘fitness cost’ (Vogwill
Microcosm study of freshly collected savannah river water and Maclean, 2015). Due to this fitness cost, the cell be-
samples, using Ralstonia mannitolilytica, an opportunis- comes biologically less efficient and only reproduces
tic human pathogen, provided experimental evidence of under selection pressure. Alternatively, in case of no selec-
co-­selection of multiantibiotic and multimetal-­resistant tion pressure, the susceptible cells will have the ability to
micro-­organisms (Stepanauskas et al., 2006). This patho- outgrow the biologically less fit resistant cells (Andersson
gen proliferated and enriched the fresh water microcosm and Hughes,  2010). Thus, under no selection pressure,
when Cd was added to it. The transfer of ARGs across the resistant cells should ideally lose the acquired MGEs
bacterial genera, primarily through conjugation in water to eliminate the cost of fitness. However, under natural
environment, was observed to increase under subinhibi- circumstances, the rate of reversibility is fairly low due to
tory concentrations of Cu and Ag nanoparticles as well as factors such as reduction in the fitness cost due to compen-
heavy metals ions, such as Cu (II), Cr (VI), Ag(I), Zn(II) sating mutations, the absence of a fitness cost altogether,
|
4064       VATS et al.

and genetic co-­selection between the gene under selection limited exposure to low concentrations of antimicrobial
and a resistance-­conferring gene. In addition to this, the agents, such as heavy metals, in the environment, clinical
maintenance of a resistant population in the environment settings and treated animals or humans, can co-­select for
has been attributed to the sub-­MIC selection which stabi- ARGs in pathogenic micro-­organisms and thus, contrib-
lizes the fitness cost of resistance. ute to the emergence, proliferation and dissemination of
A detailed study conducted by Gullberg et al.  (2011) multidrug -­esistant bacteria. Such studies highlight the
suggests that the presence of a very low antibiotic con- need to introduce new measures to control environmental
centration plays an essential role in the selection, prolif- contamination by heavy metals and restrict the redundant
eration and maintenance of existing resistant mutants in use of antibiotics in the treatment of diseases.
addition to the selection of newly evolved mutants in the
environment or infected human organs during treatment.
This minimum selective concentration (MSC), thus, can MEC HANISMS OF CO - ­S ELECTIO N
be defined as the threshold where the fitness cost of a
resistance gene is equal to the benefit of being resistant Co-­selection of heavy metals and antibiotic resistance is
(Figure 1). This concept comprises two major hypotheses. based on overlapping between mechanisms for both these
The first one being if the MSC of an antimicrobial agent is resistances, which can either be physiological overlapping
higher than its existing concentration, then an overall de- (Cross-­resistance) or genetic overlapping (Co-­resistance/
crease or eventual disappearance of the resistance genes co-­regulation) (Figure  2). These mechanisms are dis-
will be observed. Alternatively, keeping the concentration cussed in the following sections.
of the antimicrobial agent high (≥MSC) will also keep the
existing resistance genes maintained along with selecting
for de novo resistant mutants which, in due course, will CO - ­R ESISTANCE AND CROSS -­
dominate. Thus, it can be simply stated that a directly pro- RESISTANC E: MOLECULAR
portional relationship exists between the concentration MEC HANISMS FOR CO - ­S ELECT IO N
of antimicrobial agent and the selective pressure that it
imposes. The same principle will apply to heavy metals, as Co-­resistance is established when genes for two or more
they can act as antimicrobial agents as well and are widely resistance phenotypes, located on a single genetic element,
distributed in the environment and the clinical settings. are acquired by a cell under selection pressure. In such a
This concept is supported by a report that evaluated the case, the cell ends up acquiring the genes for other resist-
MSCs of various antimicrobial agents and heavy metals ance mechanisms as well. The MGEs, such as transposons,
in E. coli cells harbouring a plasmid that had resistance plasmids and integrons, which harbour the resistance
genes for multiple antibiotics and heavy metals. The MSCs genes are involved in co-­resistance and likely undergo HGT
of CuSO4 and As were observed to be 10-­fold and 140-­fold (Ghosh et al.,  2000; Gómez-­Sanz et al.,  2013). Mercury-­
lower than the MICs of the susceptible strain of E. coli resistant genes were demonstrated to be genetically linked
(Gullberg et al.,  2014). This outcome suggests that even with ARGs on plasmids as they were co-­transferred during

Sub-MIC selecon Convenonal selecon

F I G U R E 1   The concept of minimal


Growth rate

selective concentration (MSC). Selection


of resistant bacteria occurs at MSC of the
antimicrobial agent which is lower than
MSC MICsusc MICres the minimum inhibitory concentration
Concentration of the Antimicrobial agent for susceptible (MICsusc) and resistant
(MICres) cells. (adapted from Gullberg
Resistant strain Suscepble strain Dead cell
et al., 2011)
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4065

F I G U R E 2   Mechanisms behind
co-­selection of antibiotic and metal
resistance. (adapted from Baker-­Austin
et al., 2006, Pal et al., 2017)

ARG
Plasmid

HMRG

mating between Enterobacteriaceae and recipients. Ghosh vector has contributed to the unprecedented dissemina-
et al. (2000) reported the presence of plasmids in various tion of antibiotic and metal resistance since metal resis-
strains of S. abortis showing co-­resistance for ampicillin tance is majorly governed by MGEs, for example, Tn21
and multiple heavy metals, such as As, Cd, Hg and Cr. and Tn21-­like transposons harbouring Hg resistance gene
On removing these plasmids from the cell, the bacteria and ARGs. This fact was supported by a report wherein a
became sensitive to these antimicrobials proving that the conjugative plasmid, harboured by Enterococcus faecium,
genes for these agents were indeed present on plasmids. was observed to comprise of novel mercury-­resistance op-
Furthermore, in a study by Hasman and Aarestrup, the eron containing a Tnmer1 putative transposon as well as
authors reported a strong correlation between copper re- streptomycin resistance gene (aadK) (Davis et al.,  2005;
sistance and macrolide and glycopeptides resistances. In a Baker-­Austin et al., 2006).
set of experiments, they were able to co-­transfer plasmids Cross-­resistance is observed when different antimicro-
from E. facieum obtained from pigs, harbouring the tcrB bial agents employ similar modes of action to kill a mi-
and ermB genes responsible for copper and macrolides re- crobial cell. The affected cell adopts mechanisms, such
sistance, respectively, to a susceptible recipient (Hasman as multidrug efflux systems, alteration of the cell mem-
and Aarestrup, 2002). Similarly, a type of IncA/C plasmid, brane, mutations or some outer-­membrane adaptations,
harbouring a mercury resistance determinant (mer operon) that may confer resistance against more than one antimi-
and multiple ARGs was isolated from Aeromonas salmo- crobial agent (Aendekerk et al.,  2002). Cross-­resistance
nicida from aquaculture facilities (Mcintosh et al., 2008). is most commonly conferred by multidrug efflux pumps
The use of advanced sequencing and other bioinformat- which expel antibiotics and heavy metals outside the bac-
ics platforms to analyse existing data allows an improved terial cells (Martinez, 2009). The addition of vanadate to
understanding of natural co-­resistance mechanisms. The soil due to its contamination by refinery oil was reported
examination of these reports shows that genetic elements, to induce MDR in Enterobacter cloacae and Escherichia
through multiple recombination events, have acquired hermannii through a membrane-­bound efflux system
transposase and other insertion sequences (Dennis, 2005) (Hernández et al.,  1998). Many more such studies that
and might also harbour antibiotic and metal resistance report cross-­resistance between antibiotics and metals
genes. For example genetic analysis of Salmonella enteric through enhanced efflux systems are available, some of
serovar Typhi CT18 unveiled the existence of multiple an- which are listed in Table 1.
tibiotic and Hg resistance genes on a conjugative plasmid
referred to as pHCM1 (Parkhill et al., 2001).
AMR genes are most commonly incorporated into CO - R
­ EGULATION AS A
transposons which are one of the best-­documented ex- MECHANISM FOR CO - ­S ELECTI O N
amples of co-­resistance occurring through MGEs (Liebert
et al., 1999; Mindlin et al., 2001; Baker-­Austin et al., 2006). Heavy metal exposure, apart from triggering co-­
Their efficient mechanism of introducing genes to a new selection through co-­resistance and cross-­resistance,
4066       | VATS et al.

has also been reported to enhance tolerance to antibi-

Aendekerk et al. (2002)
otics by triggering co-­regulation of ARGs. Metal ex-

Hayashi et al. (2000)

Teixeira et al. (2016)
Nishino et al. (2007)
X. Z. Li et al. (1997)
Cheng et al. (1996)
posure up-­regulates the expression of ARGs, as in the

Mata et al. (2000)
Kumar (1996)

Kaur et al. (2021)
Choudhury and
case of Enterobacteriaceae bacterium LSJC7, thereby
conferring an enhanced ability to survive antibiotic
exposure. Arsenate upregulates multiple ARG emrD,
and tetracycline resistance gene tet34 in LSJC7 strain,
a Gram-­negative member of Enterobacteriaceae (Chen
et al.,  2015). The fact that this upregulation was ob-

MexGHI–­OpmD efflux pump, RND efflux


Outer membrane proteins, efflux pumps

served at environmentally relevant concentrations of


DsbA–­DsbB multidrug efflux system

AcrD and MdtABC multidrug efflux


arsenate should be a cause of perturbation. Cu or Zn
along with the BaeR response regulator induces expres-
sion of acrD and mdtABC genes which code for AcrD
Outer membrane proteins

and MdtABC multidrug efflux pumps, responsible for


Multidrug efflux systems
Multidrug efflux pumps

Multidrug efflux pumps


antibiotic resistance in Salmonella enterica serovar
Shared mechanism

Typhimurium (Nishino et al.,  2007). Similarly, the ex-


pression of SoxS, which is a universal response regulator
Efflux pumps

protein that regulates multidrug efflux pump AcrAB in


pumps

pumps

E. coli and Salmonella sp., is upregulated in response to


oxidative stress caused by ions, such as Cd, Cu2+, and
Cr2O7− (Harrison et al., 2009; Kaur et al., 2021). Cd and
As, Cd, Cr, Cu, Hg,

Cu2+ ions also act as an inducer to derepress the multiple


As, Cr, Cu, Hg, Ni

antibiotic resistance regulatory operon marRAB in both


Zn, Co, Cd

Salmonella sp. and E. coli. The membrane-­associated


Vanadium

Cadmium
Ni, Pb
Metal(s)

copper oxidation and liberation through oxidative im-


Cu, Zn
Cd, Zn
T A B L E 1   Examples of studies that have reported cross-­resistance between metals and antibiotics

pairment of copper-­dependent envelope proteins, such


Ag
Co

as NADH dehydrogenase-­2, derepress the multiple anti-


Tetracyclines, Penicillin, Chloramphenicol, Cefotaxime
Penicillin, Quinolone, Tetracycline, Trimethoprim/

Beta Lactams, Kanamycin, Erythromycin, Novobiocin,

biotic resistance regulator (MarR), leading to increased


Ticarcillin And Clavulanic Acid; Fluoroquinolones,

antibiotic resistance (Eaves et al., 2004; Hao et al., 2014).


Tetracyclines, chloramphenicol, Beta Lactams

Increased production of SoxS and MarA subsequently


Aminoglycoside, Amphenicol, Cephalosporin,

Ampicillin, ciprofloxacin, chloramphenicol,

increases the expression of AcrAB which results in tol-


Beta-­lactams, Novobiocin, Deoxycholate
Erythromycin, Josamycin, Clindamycin

erance against antibiotics, for example, tetracycline,


novobiocin, oxacillin and chloramphenicol (Figure  3)
(Harrison et al., 2009). Co-­regulation was also reported
between carbapenem and heavy metals in Pseudomonas
aeruginosa. Initially, Conejo et al.  (2003) reported that
Some Cephalosporins

the expression of a porin protein, OprD2, responsible


Sulfamethoxazole

for the influx of carbapenem antibiotics, was observed


ceftizoxime

to be downregulated in P. aeruginosa in response to Zn


Antibiotic(s)

Ofloxacin
Tetracycline

eluted from siliconized latex urinary catheters. Zn ap-


parently had a negative effect on the expression of influx
protein OprD2, thereby rendering this bacterium resist-
ant to carbapenem. Later on, heavy metal resistance due
to Zn-­induced overexpression of CzrCBA, an RND type
E. coli, Pseudomonas aeruginosa

efflux pump, and two-­component regulator genes czcR


Salmonella enterica serovar

and czcS was also found in carbapenem-­resistant strains


Pseudomonas aeruginosa

Pseudomonas aeruginosa

of P. aeruginosa (Perron et al., 2004) suggesting a com-


Lysteria monocytogenes
Burkholderia cepacia

mon regulatory mechanism between this overexpres-


Micro-­organism

Salmonella Typhi
Typhimurium

sion and the repression of OprD2.


Bacillus subtilis

Besides these co-­selection strategies, combined re-


sistance to antibiotics and metals is aided by the forma-
E. coli

tion of biofilms also. The heavy metal or antibiotic stress


induces the production of certain substances that aid
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4067

F I G U R E 3   Possible pathways for co-­regulation of heavy metal induced antibiotic resistance. Regulated by universal regulators SoxS or
MarA, through the acrAB operon that codes for the AcrAb-­TolC efflux pump. The transcription of MarA is repressed by a negative regulator,
MarR. The presence of metal ions derepresses the marRAB operon by catalysing the formation of MarR tetramer which has an attenuated
DNA-­binding ability. Gene clusters are represented by coloured arrows; * indicates the induced SoxR regulator protein (Harrison et al., 2009;
Hao et al., 2014)

in cellular adhesion along with the production of extra- VALIDATION OF CO - ­S ELECTIO N


cellular polymeric substances (EPS) in planktonic cells OF METAL AND ANTIBIOTIC
which subsequently helps in the formation of biofilms RESISTANCE IN CLINIC AL
(Baker-­Austin et al.,  2006). Induction of biofilm due to SETTINGS
production of EPS as a survival strategy on exposure to
metals has been reported by García-­Meza et al. (2005) and Abundant studies reporting the co-­selection of metal and
Kidambi et al. (1995) in Phormidium (cyanobacteria) and antibiotic resistance in various environmental contexts
Pseudomonas species respectively. Other than this, the are available but there is a paucity of studies directed
presence of bacterial biofilms in metal-­contaminated acti- solely at examining co-­selection in clinical settings. Links
vated sludge has been demonstrated by other researchers of co-­selection in a clinically relevant strain were first
as well (Wuertz et al., 2004). Likewise, the expression of established in the early 1960s (Moore, 1960). In a more
ica, an intracellular precursor for the synthesis of poly- recent bioinformatic study, through a systematic ap-
saccharide adhesins, has been shown to be promoted proach to study complete sequenced bacterial genomes
by subinhibitory concentrations of antibiotics, such and plasmids, a high ratio of plasmids was found in clin-
as tetracycline, in Staphylococcus epidermidis (Rachid ically relevant genera, including Escherichia, Klebsiella,
et al., 2000). Thus, antibiotic and metal exposure has been Staphylococcus and Salmonella (Pal et al.,  2015). The
proved to induce biofilm production in microbial commu- majority of the plasmids in these bacteria carried genes
nities, which in turn helps the bacteria to resist both these for both metal and antibiotic resistance, thus, indicat-
antimicrobial agents. ing a potential for co-­selection through co-­resistance.
|
4068       VATS et al.

This directs a pervasiveness of heavy metal-­induced resistance in clinical isolates or clinically relevant
antibiotic resistance in clinical pathogens that has not micro-­organisms.
been studied enough. Correlation between metals and A heavy metal selection pressure exists in the clinical
antibiotic resistance in micro-­organisms was validated settings, for example due to the use of surgical equipment,
in a study on clinical isolates of Salmonella enterica dental fillings, metal-­containing disinfectants, thermom-
serovar Typhi (Kaur et al.,  2018). This study observed eters or other metallic apparatus. However, direct human
that on supplementation of Cd, the sensitive isolates of exposure to heavy metals through factors such as diet
this organism were rendered resistant and the already (EFSA,  2004), or smoking (Ashraf,  2012) has also been
resistant isolates were rendered more resistant in terms reported which may co-­select for antibiotic resistance.
of their MICs of antibiotics, including ampicillin, cip- This was indicated by an analysis of the national health
rofloxacin, chloramphenicol and ceftizoxime, through and nutrition examination survey where the effects of
a co-­regulatory mechanism of co-­selection. Table  2 blood lead (Pb) levels on nasal colonization of methicillin-­
further lists some selected studies that establish a cor- resistant Staphylococcus aureus (MRSA) and methicillin-­
relation between the presence of antibiotic and metal susceptible Staphylococcus aureus (MSSA) were studied.

T A B L E 2   Examples of studies reporting a correlation between metal and antibiotic resistance in clinical settings

Clinically relevant micro-­


organism(s) exhibiting
co-­selection Antibiotic(s) Metal(s)
Staphylococcus aureus Penicillin Mercury Novick and Roth (1968)
Salmonella Typhimurium Chloramphenicol, Ampicillin Silver, mercury Larkin Mchugh
et al. (1975)
Pseudomonas aeruginosa Streptomycin, Kanamycin, Chloramphenicol Mercury, cadmium, Nakahara et al. (1977)
arsenic
Escherichia coli Streptomycin, Tetracycline, Chloramphenicol, Mercury Nakahara et al. (1977)
Kanamycin
Oral Streptococci, members of Ampicillin, Streptomycin, Tetracycline Mercury Summers et al. (1993)
Enterobacteriaceae
Stenotrophomonas maltophilia Macrolide Cadmium Alonso et al. (2000)
Staphylococcus aureus Penicillin G, Methicillin, Oxacillin Lead, chromium Ugur and Ceylan (2003)
Pseudomonas aeruginosa, Kanamycin, nalidixic acid, oxacillin, Cadmium EFSA (2004)
Klebsiella pneumoniae sulphonamides
Pseudomonas aeruginosa Carbapenem Zinc, copper Caille et al. (2007); Conejo
et al. (2003); Perron
et al. (2004)
Escherichia coli, Klebsiella Beta-­lactams, Aminoglycosides, Trimethoprim, Silver, copper, arsenic Sandegren et al. (2012)
pneumoniae Tetracyclines, Macrolides
Staphylococcus aureus Methicillin Zinc, cadmium Nair et al. (2014)
Staphylococcus aureus Amoxicillin/Clavulanic Acid, Co-­Trimoxazole, Lead Garhwal et al. (2014)
Cefoxitin, Amoxicillin/Clavulanic Acid
Members of Enterobacteriaceae Third-­ and Fourth-­generation Cephalosporins, Lead Garhwal et al. (2014)
Gentamycin, Ciprofloxacin, Co-­Trimoxazole,
Tetracycline
Salmonella Typhi Nalidixic Acid, Levofloxacin, Lead Garhwal et al. (2014)
Ampicillin-­Sulbactam
Pseudomonas sp. Colistin, Levofloxacin, Cefepime Lead Garhwal et al. (2014)
Streptococcus agalactiae Macrolide, Lincosamide Copper, cadmium Rojo-­Bezares et al. (2016)
Klebsiella pneumoniae Gentamicin, Chloramphenicol, Beta-­lactams, Mercury, copper, Zhai et al. (2016)
Rifampicin, Streptomycin, Trimethoprim, Tellurite, lead, cobalt,
Aminoglycosides cadmium
Salmonella Typhi Ampicillin, ciprofloxacin, chloramphenicol, Cadmium Kaur et al. (2018)
ceftizoxime
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4069

This study observed an increase in nasal MRSA coloniza- DPDS and the antibiotics was indicated by the fractional
tion in people with higher blood Pb levels, suggesting the inhibitory concentration (FIC) indices, which might be
selection of methicillin resistance under a selection pres- due to the multipronged action of both the agents admin-
sure posed by Pb present in the blood as one of the factors istered together.
(Eggers et al., 2018). This indicates that heavy metals have Another study suggested the use of dandelion root
a far-­reaching impact on the co-­selection of antibiotic ­extracts and taraxasterol for the reversal of metal (Cd, Ni,
resistance, so much so, that heavy metals present inside As) induced ampicillin and kanamycin resistance in E. coli
a human body may also have a role in the selection and cells (Yang and Zhang, 2020). While treatment of E. coli
propagation of antibiotic resistance. Hence, this calls for a cells with low concentrations of Dandelion root extract
thorough study of the influence of heavy metals in the se- alone did not seem to show any antimicrobial activity,
lection of antibiotic resistance, particularly in the clinical the addition of kanamycin and ampicillin along with the
context; as such scientific studies are still a rarity. heavy metals and the root extracts significantly reduced
the bacterial growth, indicating that dandelion root ex-
tracts can be effective modulators against metal-­induced
STR AT EG I E S TO DISAR M T HE antibiotic resistance.
CO - ­S E L ECT ION OF HE AVY METAL Since, antibiotic-­resistant pathogens of clinical rele-
A ND A N T I BIOT IC R E SISTAN CE vance originate from nonclinical environmental settings,
an alternate approach to disarm metal-­induced antibiotic
Since the theory of co-­selection of metal and antibiotic re- resistance can be direct in situ removal of metals from pol-
sistance has been validated through many studies, the next luted environmental sites or pretreatment of metal-­ridden
pragmatic step would be to discover strategies to curb the industrial effluents which form a conducive environment
emergence of MDR bacteria through it. Designing new for co-­selection to take place. This may be done through
therapies that use biological, chemical, or artificially de- processes such as bioremediation, biosorption, bioaccu-
signed inhibitors for pharmacological inhibition of active mulation and precipitation using algae, plants or micro-­
efflux pumps are becoming attractive strategies to combat organisms (Ahluwalia and Goyal,  2007; Parameswari
metal-­induced antibiotic resistance (Domalaon et al., 2018). et al., 2009). Several micro-­organisms have evolved various
The potency of these agents can be increased by using genetically determined mechanisms to remove toxic ions
them in combination with antibiotics so that they can be from their vicinity (Chang et al., 1997; Al-­Musharafi, 2016;
used at lower concentrations to simultaneously reduce Chellaiah, 2018). Although the use of micro-­organisms for
any chances of emergence of resistance. Pathogenicity of bioremediation has been reported as an economic alterna-
the micro-­organism is also reduced by these agents as they tive, the potential spread of heavy metal resistance deter-
have been reported to inhibit biofilm formation (Sabatini minants in the environment also needs to be considered
et al., 2017; Anes et al., 2019; Ferrer-­Espada et al., 2019). Use as these bioremedial mechanisms make the cells metal-­
of 1-­(1-­napthylmethyl)-­piperazine or NMP, one such active resistant as well. This is a major limitation of utilizing bac-
efflux pump inhibitor, was reported to reverse AcrAB ef- teria for bioremediation as exposing metal-­resistant cells
flux pump mediated MDR in cadmium adapted Salmonella to heavy metal selection pressure might select and perpet-
Typhi cells (Kaur et al., 2021). Therefore, discovering more uate heavy metal resistance markers in the environment.
such inhibitor molecules, which on administration with So, a procedure that utilizes the metal removing properties
antibiotics can reverse MDR in micro-­organisms, can be of micro-­organisms in a controlled manner, to simultane-
very beneficial for the medical community. ously restrain the dissemination of metal-­resistant bacte-
Use of a metal chelator, 2, 2′-­dipyridyl diselane ria into the environment, might be a suitable approach.
(DPDS), a chemically synthesized organoselenium com- Biosorption is a cost-­effective and relatively safer option
pound, along with antibiotics has been reported as a new as there is no risk of dissemination of metal-­resistant bac-
combinatorial approach to managing Cd-­induced antibi- teria. Dead or inactive bacteria, plant and fungus-­derived
otic resistance in clinical isolates of Salmonella enterica biomass obtained from various industrial fermentations
serovar Typhi (Rishi et al., 2017). On the treatment of Cd-­ can act as a cheap and efficient source of biosorptive ma-
adapted and antibiotic-­resistant cells of S. enterica with terial that can absorb and concentrate metal ions from the
DPDS in combination with antibiotics, such as ampicillin polluted site (Chang et al.,  1997; Issazadeh et al.,  2013;
or cefotaxime, the resistant cells were rendered sensitive Vázquez-­Núñez et al., 2018). Removal of metal ions from
to these antibiotics again. DPDS was responsible for de- polluted sites will ensure an absence of heavy metal selec-
stabilizing the structural organization of cells by chelating tion pressure for environmental microbial communities,
both essential as well as nonessential (Cd) metal ions. A thereby inhibiting co-­selection of antibiotic resistance and
synergistic and additive effect for various combinations of its subsequent transfer to pathogenic micro-­organisms.
|
4070       VATS et al.

CON C LUS I ON AN D FU T U R E CONFLICT OF INTEREST


P E R S P ECT I VE S The authors have no conflict of interest to disclose.

The challenge of antibiotic resistance, because of the ex- ORCID


tensive exploitation of antibiotics within a population, is Praveen Rishi  https://orcid.org/0000-0001-5425-8064
well recognized. Scientists, doctors, and even the general
public around the world is familiar with the implications REFERENCES
of the emergence of MDR in pathogenic bacteria. However, Aendekerk, S., Aendekerk, S., Ghysels, B., Cornelis, P. & Baysse,
selection pressure from other agents such as heavy metals C. (2002) Characterization of a new efflux pump, MexGHI-­
is also a cause of concern as they are much more prevalent OpmD, from Pseudomonas aeruginosa that confers resistance
to vanadium. Microbiology, 148(8), 2371–­2381. https://doi.
and obstinate in the environment than antibiotics. Heavy
org/10.1099/00221​287-­148-­8-­2371
metals induce antibiotic resistance in micro-­organisms by
Ahluwalia, S.S. & Goyal, D. (2007) Microbial and plant derived bio-
selecting the resistance determinants for both antibiotic mass for removal of heavy metals from wastewater. Bioresource
and metal resistance through mechanisms of co-­resistance, Technology, 98(12), 2243–­2257. https://doi.org/10.1016/j.biort​
cross-­resistance and co-­regulation. Identifying the domi- ech.2005.12.006
nant mechanism responsible for co-­selection is crucial for Al-­Musharafi, S.K. (2016) Heavy metals in sewage treated efflu-
elucidating possible targets for developing novel drugs. ents: pollution and microbial bioremediation from arid re-
The environment is the ultimate source of resistance gions. Open Biotechnology Journal, 10(1), 352–­362. https://doi.
org/10.2174/18740​70701​61001​0352
determinants, from where they are transferred to clinical
Alonso, A., Sanchez, P. & Martínez, J.L. (2000) Stenotrophomonas
settings (Alonso et al.,  2001). Reports on metal and an-
maltophilia D457R contains a cluster of genes from ­gram-­positive
tibiotic co-­selection in pathogenic bacteria are increasing bacteria involved in antibiotic and heavy metal resistance.
globally. It should be highlighted that the bacterial strains Antimicrobial Agents and Chemotherapy, 44(7), 1778–­1782.
utilized for studying co-­selection of metals and antibiotics https://doi.org/10.1128/AAC.44.7.1778-­1782.2000
in almost all the reports were isolated from the environ- Alonso, A., Sánchez, P. & Martínez, J.L. (2001) Environmental selec-
ment, indicating a lack of data on co-­selection in clinical tion of antibiotic resistance genes. Environmental Microbiology,
isolates. There is a huge prospect for studies explicitly in- 3(1), 1–­9. https://doi.org/10.1046/j.1462-­2920.2001.00161.x
Aminov, R.I. (2010) A brief history of the antibiotic era: lessons
vestigating the process of co-­selection in clinical settings
learned and challenges for the future. Frontiers in Microbiology,
and assessing its impact on mammalian disease-­causing 1(DEC), 134. https://doi.org/10.3389/fmicb.2010.00134
bacteria. Comprehensive studies analysing the environ- Andersson, D.I. & Hughes, D. (2010) Antibiotic resistance and its cost:
mental resistance gene pools and the potential of these is it possible to reverse resistance? Nature Reviews. Microbiology,
genes to transfer into pathogens of clinical significance 8(4), 260–­271. https://doi.org/10.1038/nrmic​ro2319
are also vital. The current advances in microbial genom- Anes, J., Sivasankaran, S.K., Muthappa, D.M., Fanning, S. &
ics and procedures for comprehensive genetic, transcrip- Srikumar, S. (2019) Exposure to sub-­inhibitory concentra-
tions of the chemosensitizer 1-­(1-­naphthylmethyl)-­piperazine
tional and translational analysis can form the basis for
creates membrane destabilization in multi-­drug resistant
such studies.
Klebsiella pneumoniae. Frontiers in Microbiology, 10, 92. https://
In view of rapidly proliferating antibiotic resistance doi.org/10.3389/fmicb.2019.00092
and depression in the field of discovery and develop- Ashraf, M.W. (2012) Levels of heavy metals in popular cig-
ment of newer antibiotic drugs, it is essential to look arette brands and exposure to these metals via smok-
for alternate therapeutic strategies. Since co-­selection ing. Scientific World Journal, 2012, 729430. https://doi.
of metal and antibiotic resistance is highly predominant org/10.1100/2012/729430
and adds to the burden of selection and dissemination Aslam, B., Wang, W., Arshad, M.I., Khurshid, M., Muzammil, S.,
Rasool, M. et al. (2018) Antibiotic resistance: a rundown of
of antibiotic resistance globally, it is critical to discover
a global crisis. Infection and Drug Resistance, 11, 1645–­1658.
novel therapeutics to combat it. The use of metal chela-
https://doi.org/10.2147/IDR.S173867
tors to sensitize metal-­adapted bacteria turned out to be Baker-­Austin, C., Wright, M.S., Stepanauskas, R. & McArthur, J.v.
successful in vitro, but its commercialization as an anti- (2006) Co-­selection of antibiotic and metal resistance. Trends
bacterial therapy to treat infections in vivo would be the in Microbiology, 14(4), 176–­182. https://doi.org/10.1016/j.
next bigger challenge. More such therapies and meth- tim.2006.02.006
ods need to be developed to control the metal-­induced Bass, L., Liebert, C.A., Lee, M.D., Summers, A.O., White, D.G.,
promulgation of antibiotic resistance. Reframing the Thayer, S.G. et al. (1999) Incidence and characterization of
integrons, genetic elements mediating multiple-­drug resis-
existing guidelines for the management of heavy metal-­
tance, in avian Escherichia coli. Antimicrobial Agents and
ridden waste disposal and industrial effluent treatment Chemotherapy, 43(12), 2925–­2929. https://doi.org/10.1128/
is also critical as they assist in the maintenance of ARG aac.43.12.2925
pools in the environment.
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4071

Bednorz, C., Oelgeschläger, K., Kinnemann, B., Hartmann, S., Water Research, 31(7), 1651–­1658. https://doi.org/10.1016/
Neumann, K., Pieper, R. et al. (2013) The broader context of S0043​-1­ 354(97)00008​-­0
antibiotic resistance: zinc feed supplementation of piglets in- Chellaiah, E.R. (2018) Cadmium (heavy metals) bioremediation by
creases the proportion of multi-­resistant Escherichia coli in Pseudomonas aeruginosa: a minireview. Applied Water Science,
vivo. International Journal of Medical Microbiology, 303(6–­7), 8(6), 154. https://doi.org/10.1007/s1320​1-­018-­0796-­5
396–­403. https://doi.org/10.1016/j.ijmm.2013.06.004 Chen, S., Li, X., Sun, G., Zhang, Y., Su, J. & Ye, J. (2015) Heavy metal
Benveniste, R. & Davies, J. (1973) Aminoglycoside antibiotic induced antibiotic resistance in bacterium LSJC7. International
inactivating enzymes in actinomycetes similar to those Journal of Molecular Sciences, 16(10), 23390–­23404. https://doi.
present in clinical isolates of antibiotic resistant bacteria. org/10.3390/ijms1​61023390
Proceedings of the National Academy of Sciences of the United Cheng, J., Hicks, D.B. & Krulwich, T.A. (1996) The purified Bacillus
States of America, 70(8), 2276–­2280. https://doi.org/10.1073/ subtilis tetracycline efflux protein TetA(L) reconstitutes both
pnas.70.8.2276 tetracycline-­cobalt/H+ and Na+(K+)/H+ exchange. Proceedings
Berg, J., Thorsen, M.K., Holm, P.E., Jensen, J., Nybroe, O. & of the National Academy of Sciences of the United States of
Brandt, K.K. (2010) Cu exposure under field conditions America, 93(25), 14446–­14451. https://doi.org/10.1073/
coselects for antibiotic resistance as determined by a novel pnas.93.25.14446
cultivation-­independent bacterial community tolerance assay. Choudhury, P. & Kumar, R. (1996) Association of metal tolerance
Environmental Science & Technology, 44(22), 8724–­8728. with multiple antibiotic resistance of enteropathogenic organ-
https://doi.org/10.1021/es101​798r isms isolated from coastal region of deltaic Sunderbans. The
Berg, J., Tom-­Petersen, A. & Nybroe, O. (2005) Copper amendment Indian Journal of Medical Research, 104(July), 148–­151.
of agricultural soil selects for bacterial antibiotic resistance Conejo, C.C., García, I., Martínez-­Martínez, L., Picabea, L. &
in the field. Letters in Applied Microbiology, 40(2), 146–­151. Pascual, A. (2003) Zinc eluted from siliconized latex urinary
https://doi.org/10.1111/j.1472-­765X.2004.01650.x catheters decreases OprD expression, causing carbapenem
Bhattacharya, M., Choudhury, P. & Kumar, R. (2000) Antibiotic-­ and resistance in Pseudomonas aeruginosa. Antimicrobial Agents
metal-­resistant strains of Vibrio parahaemolyticus isolated from and Chemotherapy, 47(7), 2313–­2315. https://doi.org/10.1128/
shrimps. Microbial Drug Resistance, 6(2), 171–­172. https://doi. AAC.47.7.2313-­2315.2003
org/10.1089/10766​29004​19492 D’Costa, V.M., McGrann, K.M., Hughes, D.W. & Wright, G.D. (2006)
Boyce, J.M. (2007) Environmental contamination makes an import- Sampling the antibiotic resistome. Science, 311(5759), 374–­377.
ant contribution to hospital infection. The Journal of Hospital https://doi.org/10.1126/scien​ce.1120800
Infection, 65(Suppl. 2), 50–­54. https://doi.org/10.1016/S0195​ Davis, I.J., Roberts, A. P., Ready, D., Richards, H., Wilson, M.,
-­6701(07)60015​-­2 Mullany, P. et al. (2005) Linkage of a novel mercury resistance
Boyd, E.S. & Barkay, T. (2012) The mercury resistance operon: From operon with streptomycin resistance on a conjugative plasmid
an origin in a geothermal environment to an efficient detoxifi- in enterococcus faecium. Plasmid, 54(1), 26–­38. https://doi.
cation machine. Frontiers in Microbiology, 3(Oct), 349. https:// org/10.1016/j
doi.org/10.3389/fmicb.2012.00349 Dennis, J.J. (2005) The evolution of IncP catabolic plasmids.
Burridge, L., Weis, J.S., Cabello, F., Pizarro, J. & Bostick, K. (2010) Current Opinion in Biotechnology, 16(3), 291–­298. https://doi.
Chemical use in salmon aquaculture: a review of current prac- org/10.1016/j.copbio.2005.04.002
tices and possible environmental effects. Aquaculture, 306(1–­ Ding, J., An, X.L., Lassen, S.B., Wang, H.T., Zhu, D. & Ke, X. (2019)
4), 7–­23. https://doi.org/10.1016/j.aquac​ulture.2010.05.020 Heavy metal-­induced co-­selection of antibiotic resistance genes
Caille, O., Rossier, C. & Perron, K. (2007) A copper-­activated two-­ in the gut microbiota of collembolans. Science of the Total
component system interacts with zinc and imipenem resistance Environment, 683, 210–­215. https://doi.org/10.1016/j.scito​
in Pseudomonas aeruginosa. Journal of Bacteriology, 189(13), tenv.2019.05.302
4561–­4568. https://doi.org/10.1128/JB.00095​-­07 Domalaon, R., Idowu, T., Zhanel, G. G., and Schweizer, F. (2018)
Cardoza, L.A., Knapp, C.W., Larive, C.K., Belden, J.B., Lydy, Antibiotic hybrids: the next generation of agents and adju-
M. & Graham, D.W. (2005) Factors affecting the fate of ci- vants against gram-­negative pathogens? Clinical Microbiology
profloxacin in aquatic field systems. Water, Air, and Soil Reviews 31(2), e0007643. American Society for Microbiology.
Pollution, 161(1–­4), 383–­398. https://doi.org/10.1007/s1127​ https://doi.org/10.1128/CMR.00077​-­17.
0-­005-­5550-­6 Dunne, K.A., Chaudhuri, R.R., Rossiter, A.E., Beriotto, I., Browning,
Cavaco, L.M., Hasman, H., Aarestrup, F.M., Wagenaa, J.A., D.F. et al. (2017) Sequencing a piece of history: complete ge-
Graveland, H., Veldman, K. et al. (2011) Zinc resistance of nome sequence of the original Escherichia coli strain. Microbial
Staphylococcus aureus of animal origin is strongly associated Genomics, 3(3), mgen000106. https://doi.org/10.1099/mgen.​
with methicillin resistance. Veterinary Microbiology, 150(3–­4), 0.000106
344–­348. https://doi.org/10.1016/j.vetmic.2011.02.014 Eaves, D.J., Ricci, V. & Piddock, L.J.V. (2004) Expression of acrB,
Cavaco, L.M., Hasman, H., Stegger, M., Andersen, P.S., Skov, R., acrF, acrD, marA, and soxS in salmonella enterica serovar typh-
Fluit, A.C. et al. (2010) Cloning and occurrence of czrC, a gene imurium: role in multiple antibiotic resistance. Antimicrobial
conferring cadmium and zinc resistance in methicillin-­resistant Agents and Chemotherapy, 48(4), 1145–­1150. https://doi.
Staphylococcus aureus CC398 isolates. Antimicrobial Agents org/10.1128/AAC.48.4.1145-­1150.2004
and Chemotherapy, 54(9), 3605–­3608. https://doi.org/10.1128/ EFSA. (2004) Opinion of the scientific panel on contaminants in
AAC.00058​-­10 the food chain related to cadmium as undesirable substance in
Chang, J.S., Law, R. & Chang, C.C. (1997) Biosorption of lead, copper animal feed. EFSA Journal, 2(6), 72. https://doi.org/10.2903/j.
and cadmium by biomass of Pseudomonas aeruginosa PU21. efsa.2004.72
|
4072       VATS et al.

Eggers, S., Safdar, N. & Malecki, K.M. (2018) Heavy metal exposure Gómez-­Sanz, E., Kadlec, K., Feßler, A.T., Zarazaga, M., Torres, C. &
and nasal Staphylococcus aureus colonization: analysis of the Schwarz, S. (2013) Novel erm(T)-­carrying multiresistance plas-
National Health and nutrition examination survey (NHANES). mids from porcine and human isolates of methicillin-­resistant
Environmental Health, 17(1), 2. https://doi.org/10.1186/s1294​ Staphylococcus aureus ST398 that also harbor cadmium and
0-­017-­0349-­7 copper resistance determinants. Antimicrobial Agents and
Ehrlich, H.L. (1997) Microbes and metals. Applied Microbiology and Chemotherapy, 57(7), 3275–­3282. https://doi.org/10.1128/
Biotechnology, 48(6), 687–­692. https://doi.org/10.1007/s0025​ AAC.00171​-­13
30051116 Gullberg, E., Albrecht, L.M., Karlsson, C., Sandegren, L. &
Essa, A.M.M., Julian, D.J., Kidd, S.P., Brown, N.L. & Hobman, Andersson, D.I. (2014) Selection of a multidrug resistance plas-
J.L. (2003) Mercury resistance determinants related to Tn21, mid by sublethal levels of antibiotics and heavy metals. MBio,
Tn1696, and Tn5053 in enterobacteria from the preantibiotic 5(5), e01918-­14. https://doi.org/10.1128/mBio.01918​-1­ 4
era. Antimicrobial Agents and Chemotherapy, 47(3), 1115–­1119. Gullberg, E., Cao, S., Berg, O.G., Ilbäck, C., Sandegren, L., Hughes,
https://doi.org/10.1128/AAC.47.3.1115-­1119.2003 D. et al. (2011) Selection of resistant bacteria at very low anti-
Fajardo, A. & Martínez, J.L. (2008) Antibiotics as signals that trigger biotic concentrations. PLoS Pathogens, 7(7), e1002158. https://
specific bacterial responses. Current Opinion in Microbiology, doi.org/10.1371/journ​al.ppat.1002158
11(2), 161–­167. https://doi.org/10.1016/j.mib.2008.02.006 Gupta, A., Matsui, K., Lo, J.F. & Silver, S. (1999) Molecular basis for
Ferrer-­Espada, R., Shahrour, H., Pitts, B., Stewart, P.S., Sánchez-­ resistance to silver cations in salmonella. Nature Medicine, 5(2),
Gómez, S. & Martínez-­de-­Tejada, G. (2019) A permeability-­ 183–­188. https://doi.org/10.1038/5545
increasing drug synergizes with bacterial efflux pump inhib- Hao, Z., Lou, H., Zhu, R., Zhu, J., Zhang, D. et al. (2014) The mul-
itors and restores susceptibility to antibiotics in multi-­drug tiple antibiotic resistance regulator MarR is a copper sensor in
resistant Pseudomonas aeruginosa strains. Scientific Reports, Escherichia coli. Nature Chemical Biology, 10(1), 21–­28. https://
9(1), 1–­12. https://doi.org/10.1038/s4159​8-­019-­39659​-­4 doi.org/10.1038/nchem​bio.1380
Flach, C.F., Pal, C., Svensson, C.J., Kristiansson, E., Östman, M., Harrison, J.J., Tremaroli, V., Stan, M.A., Chan, C.S., Vacchi-­Suzzi,
Bengtsson-­Palme, J. et al. (2017) Does antifouling paint select C. et al. (2009) Chromosomal antioxidant genes have metal
for antibiotic resistance? Science of the Total Environment, 590–­ ion-­specific roles as determinants of bacterial metal tolerance.
591, 461–­468. https://doi.org/10.1016/j.scito​tenv.2017.01.213 Environmental Microbiology, 11(10), 2491–­2509. https://doi.
Forsberg, K.J., Reyes, A., Wang, B., Selleck, E.M., Sommer, M.O.A. org/10.1111/j.1462-­2920.2009.01973.x
& Dantas, G. (2012) The shared antibiotic resistome of soil Hasman, H. & Aarestrup, F.M. (2002) Tcrb, a gene conferring trans-
bacteria and human pathogens. Science, 337(6098), 1107–­1111. ferable copper resistance in Enterococcus faecium: Occurrence,
https://doi.org/10.1126/scien​ce.1220761 transferability, and linkage to macrolide and glycopeptide re-
Fraser, C.M., Gocayne, J.D., White, O., Adams, M.D., Clayton, R.A., sistance, Antimicrob Agents. Chemotherapy, 46(5), 1410–­1416.
Fleischmann, R.D. et al. (1995) The minimal gene complement https://doi.org/10.1128/AAC.46.5.1410-­1416.2002
of mycoplasma genitalium. Science, 270(5235), 397–­403. https:// Hau, S.J., Frana, T., Sun, J., Davies, P.R. & Nicholson, T.L. (2017) Zinc
doi.org/10.1126/scien​ce.270.5235.397 resistance within swine-­associated methicillin-­resistant staph-
Fru, E.C., RodrÃ-­Guez, N.P., Partin, C.A., Lalonde, S.v., Andersson, ylococcus aureus isolates in the United States is associated with
P. et al. (2016) Cu isotopes in marine black shales record the multilocus sequence type lineage. Applied and Environmental
great oxidation event. Proceedings of the National Academy of Microbiology, 83(15), e00756–­17. http://dx.doi.org/10.1128/
Sciences of the United States of America, 113(18), 4941–­4946. aem.00756​-­17
https://doi.org/10.1073/pnas.15235​44113 Hayashi, S., Mitsuko, A., Kimoto, M., Furukawa, S. & Nakazawa,
García-­Meza, J.V., Barrangue, C. & Admiraal, W. (2005) Biofilm T. (2000) The DsbA-­DsbB disulfide bond formation system of
formation by algae as a mechanism for surviving on mine tail- Burkholderia cepacia is involved in the production of protease
ings. Environmental Toxicology and Chemistry, 24(3), 573–­581. and alkaline phosphatase, motility, metal resistance, and multi-­
https://doi.org/10.1897/04-­064R.1 drug resistance. Microbiology and Immunology, 44(1), 41–­50.
Garhwal, D., Vaghela, G., Panwala, T., Revdiwala, S., Shah, A. & https://doi.org/10.1111/j.1348-­0421.2000.tb012​44.x
Mulla, S. (2014) Lead tolerance capacity of clinical bacte- Hedges, R.W. & Baumberg, S. (1973) Resistance to arsenic com-
rial isolates and change in their antibiotic susceptibility pat- pounds conferred by a plasmid transmissible between strains
tern after exposure to a heavy metal. International Journal of of Escherichia coli. Journal of Bacteriology, 115(1), 459–­460.
Medicine and Public Health, 4(3), 253. https://doi.org/10.4103 https://doi.org/10.1128/jb.115.1.459-­460.1973
/2230-­8598.137711 Hernández, A., Mellado, R.P. & Martínez, J.L. (1998) Metal accu-
Ghosh, A., Singh, A., Ramteke, P.W. & Singh, V.P. (2000) mulation and vanadium-­induced multidrug resistance by en-
Characterization of large plasmids encoding resistance to toxic vironmental isolates of Escherichia hermannii and Enterobacter
heavy metals in Salmonella abortus equi. Biochemical and cloacae. Applied and Environmental Microbiology, 64(11),
Biophysical Research Communications, 272(1), 6–­11. https:// 4317–­4320. https://doi.org/10.1128/aem.64.11.4317-­4320.1998
doi.org/10.1006/bbrc.2000.2727 Hobman, J.L. & Crossman, L.C. (2015) Bacterial antimicrobial metal
Gillespie, S.H. (2001) Antibiotic resistance in the absence of selective ion resistance. Journal of Medical Microbiology, 64(Pt 5), 471–­
pressure. International Journal of Antimicrobial Agents, 17(3), 497. https://doi.org/10.1099/jmm.0.02303​6-­0
171–­176. https://doi.org/10.1016/S0924​-­8579(00)00340​-­X Holman, D.B. & Chénier, M.R. (2015) Antimicrobial use in swine
Gilliver, M.A., Bennett, M., Begon, M., Hazel, S.M. & Hart, C.A. production and its effect on the swine gut microbiota and anti-
(1999) Antibiotic resistance found in wild rodents. Nature, microbial resistance. Canadian Journal of Microbiology, 61(11),
401(6750), 233–­234. https://doi.org/10.1038/45724 785–­798. https://doi.org/10.1139/cjm-­2015-­0239
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4073

Hölzel, C.S., Müller, C., Harms, K.S., Mikolajewski, S., Schäfer, Lehtinen, S., Blanquart, F., Lipsitch, M. & Fraser, C. (2019) On the
S., Schwaiger, K. et al. (2012) Heavy metals in liquid pig evolutionary ecology of multidrug resistance in bacteria. PLoS
manure in light of bacterial antimicrobial resistance. Pathogens, 15(5), e1007763. https://doi.org/10.1371/journ​
Environmental Research, 113, 21–­27. https://doi.org/10.1016/j. al.ppat.1007763
envres.2012.01.002 Li, X., Gu, A.Z., Zhang, Y., Xie, B., Li, D. & Chen, J. (2019) Sub-­lethal
Hu, H.W., Wang, J.T., Li, J., Shi, X.Z., Ma, Y.B., Chen, D. et al. (2017) concentrations of heavy metals induce antibiotic resistance
Long-­term nickel contamination increases the occurrence of via mutagenesis. Journal of Hazardous Materials, 369, 9–­16.
antibiotic resistance genes in agricultural soils. Environmental https://doi.org/10.1016/j.jhazm​at.2019.02.006
Science & Technology, 51(2), 790–­800. https://doi.org/10.1021/ Li, X.Z., Nikaido, H. & Williams, K.E. (1997) Silver-­resistant mutants
acs.est.6b03383 of Escherichia coli display active efflux of ag+ and are deficient
Issazadeh, K., Jahanpour, N., Pourghorbanali, F., Raeisi, G. & in porins. Journal of Bacteriology, 179(19), 6127–­6132. https://
Faekhondeh, J. (2013) Heavy metals resistance by bacte- doi.org/10.1128/jb.179.19.6127-­6132.1997
rial strains. Scholars Research Library Annuals of Biological Li, J., Phulpoto, I.A., Zhang, G. & Yu, Z. (2021) Acceleration of emer-
Research, 4(2), 60–­63. gence of E. coli antibiotic resistance in a simulated sublethal
Jackson, C.R. & Dugas, S.L. (2003) Phylogenetic analysis of bacterial concentration of copper and tetracycline co-­contaminated en-
and archaeal arsC gene sequences suggests an ancient, com- vironment. AMB Express, 11(1), 1–­11. https://doi.org/10.1186/
mon origin for arsenate reductase. BMC Evolutionary Biology, s1356​8-­020-­01173​-6­
3(1), 1–­10. https://doi.org/10.1186/1471-­2148-­3-­18 Liebert, C.A., Hall, R.M. & Summers, A.O. (1999) Transposon Tn 21,
Kaufmann, G.F. et al. (2005) Revisiting quorum sensing: dis- flagship of the floating genome. Microbiology and Molecular
covery of additional chemical and biological functions for Biology Reviews, 63(3), 507–­522. https://doi.org/10.1128/
3-­oxo-­N-­acylhomoserine lactones. Proceedings of the National mmbr.63.3.507-­522.1999
Academy of Sciences of the United States of America, 102(2), Lu, J., Wang, Y., Jin, M., Yuan, Z., Bond, P. & Guo, J. (2020) Both
309–­314. https://doi.org/10.1073/pnas.04086​39102 silver ions and silver nanoparticles facilitate the horizontal
Kaur, U.J., Chopra, A., Preet, S., Raj, K., Kondepudi, K.K., Gupta, transfer of plasmid-­mediated antibiotic resistance genes.
V. et al. (2021) Potential of 1-­(1-­napthylmethyl)-­piperazine, an Water Research, 169, 115229. https://doi.org/10.1016/j.
efflux pump inhibitor against cadmium-­induced multidrug re- watres.2019.115229
sistance in salmonella enterica serovar typhi as an adjunct to Martinez, J.L. (2009) The role of natural environments in the evolu-
antibiotics. Brazilian Journal of Microbiology, 52, 1303–­1313. tion of resistance traits in pathogenic bacteria. Proceedings of
https://doi.org/10.1007/s4277​0-­021-­00492​-­5 the Royal Society B: Biological Sciences, 276(1667), 2521–­2530.
Kaur, U.J., Preet, S. & Rishi, P. (2018) Augmented antibiotic resis- https://doi.org/10.1098/rspb.2009.0320
tance associated with cadmium induced alterations in sal- Masindi, V. & Muedi, K.L. (2018) Environmental contamina-
monella enterica serovar typhi. Scientific Reports, 8(1), 12818. tion by heavy metals. Heavy Metal, 10, 115–­132. https://doi.
https://doi.org/10.1038/s4159​8-­018-­31143​-­9 org/10.5772/intec​hopen.76082
Kholodii, G., Mindlin, S., Petrova, M. & Minakhina, S. (2003) Mata, M.T., Baquero, F. & Pérez-­Díaz, J.C. (2000) A multidrug ef-
Tn5060 from the Siberian permafrost is most closely related flux transporter in Listeria monocytogenes. FEMS Microbiology
to the ancestor of Tn21 prior to integron acquisition. FEMS Letters, 187(2), 185–­188. https://doi.org/10.1016/S0378​
Microbiology Letters, 226(2), 251–­255. https://doi.org/10.1016/ -­1097(00)00199​-­3
S0378​-­1097(03)00559​-­7 Maurya, A.P., Rajkumari, J., Bhattacharjee, A. & Pandey, P. (2020)
Kidambi, S.P., Sundin, G.W., Palmer, D.A., Chakrabarty, A.M. & Development, spread and persistence of antibiotic resistance
Bender, C.L. (1995) Copper as a signal for alginate synthesis in genes (ARGs) in the soil microbiomes through co-­selection.
pseudomonas syringae pv. Syringae. Applied and Environmental Reviews on Environmental Health, 35(4), 371–­378. https://doi.
Microbiology, 61(6), 2172–­2179. https://doi.org/10.1128/ org/10.1515/reveh​-­2020-­0035
aem.61.6.2172-­2179.1995 Mcintosh, D., Cunningham, M., Ji, B., Fekete, F.A., Parry, E.M. et al.
Kolpin, D.W., Furlong, E.T., Meyer, M.T., Thurman, E.M., Zaugg, S.D., (2008) Transferable, multiple antibiotic and mercury resistance
Barber, L.B. et al. (2002) Pharmaceuticals, hormones, and other in Atlantic Canadian isolates of Aeromonas salmonicida subsp.
organic wastewater contaminants in U.S. streams, 1999–­2000: a salmonicida is associated with carriage of an IncA/C plasmid
national reconnaissance. Environmental Science & Technology, similar to the Salmonella enterica plasmid pSN254. The Journal
36(6), 1202–­1211. https://doi.org/10.1021/es011​055j of Antimicrobial Chemotherapy, 61(6), 1221–­1228. https://doi.
Kotlarska, E., Łuczkiewicz, A., Pisowacka, M. & Burzyński, A. org/10.1093/jac/dkn123
(2015) Antibiotic resistance and prevalence of class 1 and 2 Mighall, T.M., Abrahams, P.W., Grattan, J.P., Hayes, D., Timberlake,
integrons in Escherichia coli isolated from two wastewater S. & Forsyth, S. (2002) Geochemical evidence for atmospheric
treatment plants, and their receiving waters (gulf of Gdansk, pollution derived from prehistoric copper mining at Copa Hill,
Baltic Sea, Poland). Environmental Science and Pollution Cwmystwyth, mid-­Wales, UK. Science of the Total Environment,
Research, 22(3), 2018–­2030. https://doi.org/10.1007/s1135​ 292(1–­2), 69–­80. https://doi.org/10.1016/S0048​-­9697(02)00027​-­X
6-­014-­3474-­7 Mindlin, S. et al. (2001) Mercury resistance transposons of
Larkin Mchugh, G., Moellering, R.C., Hopkins, C.C. & Swartz, M.N. gram-­negative environmental bacteria and their classifica-
(1975) Salmonella typhimurium resistant to silver nitrate, tion. Research in Microbiology, 152(9), 811–­822. https://doi.
chloramphenicol, and ampicillin: a new threat in burn units? org/10.1016/S0923​-2­ 508(01)01265​-­7
Lancet, 305(7901), 235–­240. https://doi.org/10.1016/S0140​ Moore, B. (1960) A new screen test and selective medium for
-­6736(75)91138​-­1 the rapid detection of epidemic strains of Staph. Aureus.
|
4074       VATS et al.

Lancet, 276(7148), 453–­458. https://doi.org/10.1016/S0140​ Research, 98(1), 100–­103. https://doi.org/10.1016/j.


-­6736(60)91591​-­9 envres.2004.05.012
Nair, R., Thapaliya, D., Su, Y. & Smith, T.C. (2014) Resistance to zinc Pazda, M., Kumirska, J., Stepnowski, P. & Mulkiewicz, E. (2019)
and cadmium in Staphylococcus aureus of human and animal Antibiotic resistance genes identified in wastewater treatment
origin. Infection Control and Hospital Epidemiology, 35(S3), plant systems—­a review. Science of the Total Environment, 697,
S32–­S39. https://doi.org/10.1086/677834 134023. https://doi.org/10.1016/j.scito​tenv.2019.134023
Nakahara, H., Ishikawa, T., Sarai, Y., Kondo, I. & Kozukue, H. Peltier, E., Vincent, J., Finn, C. & Graham, D.W. (2010) Zinc-­
(1977) Mercury resistance and R plasmids in Escherichia coli induced antibiotic resistance in activated sludge bioreactors.
isolated from clinical lesions in Japan. Antimicrobial Agents Water Research, 44(13), 3829–­3836. https://doi.org/10.1016/j.
and Chemotherapy, 11(6), 999–­1003. https://doi.org/10.1128/ watres.2010.04.041
AAC.11.6.999 Perron, K., Caille, O., Rossier, C., van Delden, C., Dumas, J.L. &
Nakahara, H., Ishikawa, T., Sarai, Y., Kondo, I., Kozukue, H. & Köhler, T. (2004) CzcR-­CzcS, a two-­component system involved
Silver, S. (1977) Linkage of mercury, cadmium, and arsenate in heavy metal and carbapenem resistance in Pseudomonas
and drug resistance in clinical isolates of Pseudomonas aerugi- aeruginosa. The Journal of Biological Chemistry, 279(10), 8761–­
nosa. Applied and Environmental Microbiology, 33(4), 975–­976. 8768. https://doi.org/10.1074/jbc.M3120​80200
https://doi.org/10.1128/aem.33.4.975-­976.1977 Pietsch, F., O’Neill, A.J., Ivask, A., Jenssen, H., Inkinen, J., Kahru, A.
Nies, D.H. (1999) Microbial heavy-­metal resistance. Applied et al. (2020) Selection of resistance by antimicrobial coatings in
Microbiology and Biotechnology, 51(6), 730–­750. https://doi. the healthcare setting. The Journal of Hospital Infection, 106(1),
org/10.1007/s0025​30051457 115–­125. https://doi.org/10.1016/j.jhin.2020.06.006
Nishino, K., Nikaido, E. & Yamaguchi, A. (2007) Regulation of Poirel, L., Héritier, C. & Nordmann, P. (2004) Chromosome-­encoded
multidrug efflux systems involved in multidrug and metal re- ambler class D β-­lactamase of Shewanella oneidensis as a pro-
sistance of salmonella enterica serovar typhimurium. Journal genitor of carbapenem-­hydrolyzing oxacillinase. Antimicrobial
of Bacteriology, 189(24), 9066–­9075. https://doi.org/10.1128/ Agents and Chemotherapy, 48(1), 348–­351. https://doi.
JB.01045​-­07 org/10.1128/AAC.48.1.348-­351.2004
Novick, R.P. & Roth, C. (1968) Plasmid-­linked resistance to inorganic Poirel, L., Rodriguez-­Martinez, J.M., Mammeri, H., Liard, A. &
salts in Staphylococcus aureus. Journal of Bacteriology, 95(4), Nordmann, P. (2005) Origin of plasmid-­mediated quino-
1335–­1342. https://doi.org/10.1128/jb.95.4.1335-­1342.1968 lone resistance determinant QnrA. Antimicrobial Agents and
OÖsterblad, M., Norrdahl, K., Korpimaäki, E. & Huovinen, P. Chemotherapy, 49(8), 3523–­3525. https://doi.org/10.1128/
(2001) How wild are wild mammals? Nature, 409(6816), 37–­38. AAC.49.8.3523-­3525.2005
https://doi.org/10.1038/35051173 Raab, A. & Feldmann, J. (2003) Microbial transformation of metals
Oyetibo, G.O., Ilori, M.O., Adebusoye, S.A., Obayori, O.S. & and metalloids. Science Progress, 86(3), 179–­202. https://doi.
Amund, O.O. (2010) Bacteria with dual resistance to el- org/10.3184/00368​50037​83238671
evated concentrations of heavy metals and antibiotics in Rachid, S., Ohlsen, K., Witte, W., Hacker, J. & Ziebuhr, W. (2000)
Nigerian contaminated systems. Environmental Monitoring Effect of subinhibitory antibiotic concentrations on polysac-
and Assessment, 168(1–­4), 305–­314. https://doi.org/10.1007/ charide intercellular adhesin expression in biofilm-­forming
s1066​1-­009-­1114-­3 Staphylococcus epidermidis. Antimicrobial Agents and
Page, K., Wilson, M. & Parkin, I.P. (2009) Antimicrobial surfaces Chemotherapy, 44(12), 3357–­3363. https://doi.org/10.1128/
and their potential in reducing the role of the inanimate en- AAC.44.12.3357-­3363.2000
vironment in the incidence of hospital-­acquired infections. Resende, J.A., Silva, V.L., Fontes, C.O., Souza-­Filho, J.A., de Oliveira,
Journal of Materials Chemistry, 19(23), 3818–­3831. https://doi. T.L.R. et al. (2012) Multidrug-­resistance and toxic metal toler-
org/10.1039/b818698g ance of medically important bacteria isolated from an aqua-
Pal, C., Asiani, K., Arya, S., Rensing, C., Stekel, D.J., Larsson, D.J. culture system. Microbes and Environments, 27(4), 449–­455.
et al. (2017) Metal resistance and its association with antibi- https://doi.org/10.1264/jsme2.ME12049
otic resistance. Advances in Microbial Physiology, 70, 261–­313. Richmond, M.H. & John, M. (1964) Co-­transduction by a staphylo-
https://doi.org/10.1016/bs.ampbs.2017.02.001 coccal phage of the genes responsible for penicillinase synthesis
Pal, C., Bengtsson-­Palme, J., Kristiansson, E. & Larsson, D.G.J. and resistance to mercury salts. Nature, 202(4939), 1360–­1361.
(2015) Co-­occurrence of resistance genes to antibiotics, bio- https://doi.org/10.1038/20213​60a0
cides and metals reveals novel insights into their co-­selection Rishi, P., Thakur, R., Kaur, U.J., Singh, H. & Bhasin, K.K. (2017)
potential. BMC Genomics, 16(1), 964. https://doi.org/10.1186/ Potential of 2, 2′-­dipyridyl diselane as an adjunct to antibiotics
s1286​4-­015-­2153-­5 to manage cadmium-­induced antibiotic resistance in salmo-
Parameswari, E., Lakshmanan, A. & Thilagavathi, T. (2009) nella enterica serovar typhi Ty2 strain. Journal of Microbiology,
Biosorption of chromium (VI) and nickel (II) by bacte- 55(9), 737–­744. https://doi.org/10.1007/s1227​5-­017-­7040-­0
rial isolates from an aqueous solution. Electronic Journal of Rojo-­Bezares, B., Azcona-­Gutiérrez, J.M., Martin, C., Jareño, M.S.,
Environmental, Agricultural and Food Chemistry, 8(3), 150–­156. Torres, C. & Sáenz, Y. (2016) Streptococcus agalactiae from
Parkhill, J., Dougan, G., James, K.D., Thomson, N.R., Pickard, D. pregnant women: antibiotic and heavy-­metal resistance mech-
et al. (2001) Complete genome sequence of a multiple drug anisms and molecular typing. Epidemiology and Infection,
resistant Salmonella enterica serovar Typhi CT18. Nature, 144(15), 3205–­3214. https://doi.org/10.1017/S0950​26881​
413(6858), 848–­852. https://doi.org/10.1038/35101607 6001692
Pathak, S.P. & Gopal, K. (2005) Occurrence of antibiotic and metal Rolland, R.M., Hausfater, G., Marshall, B. & Levy, S.B. (1985)
resistance in bacteria from organs of river fish. Environmental Antibiotic-­resistant bacteria in wild primates: increased
METAL-­INDUCED ANTIBIOTIC RESISTANCE      |  4075

prevalence in baboons feeding on human refuse. Applied Environmental Microbiology, 8(9), 1510–­1514. https://doi.
and Environmental Microbiology, 49(4), 791–­794. https://doi. org/10.1111/j.1462-­2920.2006.01091.x
org/10.1128/aem.49.4.791-­794.1985 Stepanauskas, R., Glenn, T.C., Jagoe, C.H., Tuckfield, R.C., Lindell,
Romero, J.L., Grande Burgos, M.J., Pérez-­Pulido, R., Gálvez, A. & A.H. & McArthur, J.v. (2005) Elevated microbial tolerance to
Lucas, R. (2017) Resistance to antibiotics, biocides, preserva- metals and antibiotics in metal-­contaminated industrial envi-
tives and metals in bacteria isolated from seafoods: co-­selection ronments. Environmental Science & Technology, 39(10), 3671–­
of strains resistant or tolerant to different classes of com- 3678. https://doi.org/10.1021/es048​468f
pounds. Frontiers in Microbiology, 8. https://doi.org/10.3389/ Summers, A.O., Wireman, J., Vimy, M.J., Lorscheider, F.L., Marshall,
fmicb.2017.01650 B. et al. (1993) Mercury released from dental ‘silver’ fillings pro-
Rouch, D.A., Lee, B.T.O. & Morby, A.P. (1995) Understanding cel- vokes an increase in mercury-­ and antibiotic-­resistant bacteria
lular responses to toxic agents: a model for mechanism-­choice in oral and intestinal floras of primates. Antimicrobial Agents
in bacterial metal resistance. Journal of Industrial Microbiology, and Chemotherapy, 37(4), 825–­834. https://doi.org/10.1128/
14(2), 132–­141. https://doi.org/10.1007/BF015​69895 AAC.37.4.825
Rutgersson, C., Fick, J., Marathe, N., Kristiansson, E., Janzon, A. et al. Sundqvist, M., Geli, P., Andersson, D.I., Sjölund-­Karlsson, M.,
(2014) Fluoroquinolones and qnr genes in sediment, water, soil, Runehagen, A. et al. (2009) Little evidence for reversibility of
and human fecal flora in an environment polluted by manufac- trimethoprim resistance after a drastic reduction in trimetho-
turing discharges. Environmental Science & Technology, 48(14), prim use. The Journal of Antimicrobial Chemotherapy, 65(2),
7825–­7832. https://doi.org/10.1021/es501​452a 350–­360. https://doi.org/10.1093/jac/dkp387
Rwego, I.B., Gillespie, T.R., Isabirye-­Basuta, G. & Goldberg, T.L. Teixeira, P., Tacão, M., Alves, A. & Henriques, I. (2016) Antibiotic
(2008) High rates of Escherichia coli transmission between and metal resistance in a ST395 Pseudomonas aeruginosa en-
livestock and humans in rural Uganda. Journal of Clinical vironmental isolate: a genomics approach. Marine Pollution
Microbiology, 46(10), 3187–­3191. https://doi.org/10.1128/JCM.​ Bulletin, 110(1), 75–­81. https://doi.org/10.1016/j.marpo​lbul.​
00285​-­08 2016.06.086
Sabatini, S., Piccioni, M., Felicetti, T., de Marco, S., Manfroni, G. Timoney, J.F., Port, J., Giles, J. & Spanier, J. (1978) Heavy-­metal and
et al. (2017) Investigation on the effect of known potent: S. antibiotic resistance in the bacterial flora of sediments of New
aureus NorA efflux pump inhibitors on the staphylococcal bio- York bight. Applied and Environmental Microbiology, 36(3),
film formation. RSC Advances, 7(59), 37007–­37014. https://doi. 465–­472. https://doi.org/10.1128/aem.36.3.465-­472.1978
org/10.1039/c7ra0​3859c Ugur, A. & Ceylan, Ö. (2003) Occurrence of resistance to antibiot-
Sandegren, L., Linkevicius, M., Lytsy, B., Melhus, Å. & Andersson, ics, metals, and plasmids in clinical strains of staphylococcus
D.I. (2012) Transfer of an Escherichia coli ST131 multiresis- spp. Archives of Medical Research, 34(2), 130–­136. https://doi.
tance cassette has created a Klebsiella pneumoniae-­specific org/10.1016/S0188​-­4409(03)00006​-­7
plasmid associated with a major nosocomial outbreak. The Vázquez-­Núñez, E. et al. (2018) A review on genetically modified
Journal of Antimicrobial Chemotherapy, 67(1), 74–­83. https:// plants designed to Phytoremediate polluted soils: biochemical
doi.org/10.1093/jac/dkr405 responses and international regulation. Pedosphere, 28(5), 697–­
Seiler, C. & Berendonk, T.U. (2012) Heavy metal driven co-­selection 712. https://doi.org/10.1016/S1002​-­0160(18)60039​-6­
of antibiotic resistance in soil and water bodies impacted by Vogwill, T. & Maclean, R.C. (2015) The genetic basis of the fit-
agriculture and aquaculture. Frontiers in Microbiology, 3(Dec), ness costs of antimicrobial resistance: a meta-­analysis ap-
399. https://doi.org/10.3389/fmicb.2012.00399 proach. Evolutionary Applications, 8(3), 284–­295. https://doi.
Silva, I., Tacão, M. & Henriques, I. (2021) Selection of antibi- org/10.1111/eva.12202
otic resistance by metals in a riverine bacterial community. Wang, Q., Liu, L., Hou, Z., Wang, L., Ma, D. et al. (2020) Heavy
Chemosphere, 263, 127936. https://doi.org/10.1016/j.chemo​ metal copper accelerates the conjugative transfer of antibi-
sphere.2020.127936 otic resistance genes in freshwater microcosms. Science of the
Skurnik, D., Ruimy, R., Ready, D., Ruppe, E., Bernède-­Bauduin, Total Environment, 717, 137055. https://doi.org/10.1016/j.scito​
C. et al. (2010) Is exposure to mercury a driving force for the tenv.2020.137055
carriage of antibiotic resistance genes? Journal of Medical Whitman, W.B., Coleman, D.C. & Wiebe, W.J. (1998) Prokaryotes:
Microbiology, 59(7), 804–­807. https://doi.org/10.1099/ the unseen majority. Proceedings of the National Academy of
jmm.0.01766​5-­0 Sciences of the United States of America, 95(12), 6578–­6583.
Skurnik, D., Skurnik, D., Ruimy, R., Andremont, A., Amorin, C., https://doi.org/10.1073/pnas.95.12.6578
Rouquet, P. et al. (2006) Effect of human vicinity on antimicro- WHO (2014) WHO’s first global report on antibiotic resistance re-
bial resistance and integrons in animal faecal Escherichia coli. veals serious, worldwide threat to public health. [News release].
The Journal of Antimicrobial Chemotherapy, 57(6), 1215–­1219. Available at: https://www.who.int/news/item/30-­04-­2014-­
https://doi.org/10.1093/jac/dkl122 who-­s-­first​-­globa​l-­repor​t-­on-­antib​iotic​-­resis​tance​-­revea​ls-­serio​
Slifierz, M.J., Friendship, R. & Weese, J.S. (2015) Zinc oxide therapy us-­world​wide-­threa​t-­to-­publi​c-­health (Accessed: August 1,
increases prevalence and persistence of methicillin-­resistant 2021).
staphylococcus aureus in pigs: a randomized controlled trial. Wright, G.D. (2007) The antibiotic resistome: the nexus of chemical
Zoonoses and Public Health, 62(4), 301–­308. https://doi. and genetic diversity. Nature Reviews. Microbiology, 5(3), 175–­
org/10.1111/zph.12150 186. https://doi.org/10.1038/nrmic​ro1614
Stepanauskas, R., Glenn, T.C., Jagoe, C.H., Tuckfield, R.C., Lindell, Wright, G.D. (2010) Antibiotic resistance in the environment: a link
A.H., King, C.J. et al. (2006) Coselection for microbial resis- to the clinic? Current Opinion in Microbiology, 13(5), 589–­594.
tance to metals and antibiotics in freshwater microcosms. https://doi.org/10.1016/j.mib.2010.08.005
|
4076       VATS et al.

Wuertz, S., Okabe, S. & Hausner, M. (2004) Microbial communi- Zhang, X.X., Zhang, T., Zhang, M., Fang, H.H.P. & Cheng, S.P. (2009)
ties and their interactions in biofilm systems: an overview. Characterization and quantification of class 1 integrons and
Water Science and Technology, 49(11–­12), 327–­336. https://doi. associated gene cassettes in sewage treatment plants. Applied
org/10.2166/wst.2004.0873 Microbiology and Biotechnology, 82(6), 1169–­1177. https://doi.
Xiong, W., Sun, Y., Ding, X., Zhang, Y. & Zeng, Z. (2014) Antibiotic org/10.1007/s0025​3-­009-­1886-­y
resistance genes occurrence and bacterial community compo- Zhang, X.X., Zhang, T., Zhang, M., Fang, H.H.P. & Cheng, S.P. (2018)
sition in the Liuxi River. Frontiers in Environmental Science, Sub-­inhibitory concentrations of heavy metals facilitate the
2(Dec), 61. https://doi.org/10.3389/fenvs.2014.00061 horizontal transfer of plasmid-­mediated antibiotic resistance
Yang, K. & Zhang, Y. (2020) Reversal of heavy metal-­induced an- genes in water environment. Environmental Pollution, 237, 74–­
tibiotic resistance by dandelion root extracts and taraxasterol. 82. https://doi.org/10.1016/j.envpol.2018.01.032
Journal of Medical Microbiology, 69(8), 1049–­1061. https://doi. Zhou, B., Wang, C., Zhao, Q., Wang, Y., Huo, M., Wang, J. et al. (2016)
org/10.1099/jmm.0.001226 Prevalence and dissemination of antibiotic resistance genes
Yazdankhah, S., Rudi, K. & Bernhoft, A. (2014) Zinc and cop- and coselection of heavy metals in Chinese dairy farms. Journal
per in animal feed—­development of resistance and co-­ of Hazardous Materials, 320, 10–­17. https://doi.org/10.1016/j.
resistance to antimicrobial agents in bacteria of animal origin. jhazm​at.2016.08.007
Microbial Ecology in Health and Disease, 25, 25862. https://doi. Zou, H.Y., He, L.Y., Gao, F.Z., Zhang, M., Chen, S. et al. (2021)
org/10.3402/mehd.v25.25862 Antibiotic resistance genes in surface water and groundwa-
Yim, G., Huimi Wang, H. & Davies, J. (2006) The truth about antibi- ter from mining affected environments. Science of the Total
otics. International Journal of Medical Microbiology, 296(2-­3), Environment, 772, 145516. https://doi.org/10.1016/j.scito​
163–­170. https://doi.org/10.1016/j.ijmm.2006.01.039 tenv.2021.145516
Zhai, Y., He, Z., Kang, Y., Yu, H., Wang, J. et al. (2016) Complete Zouboulis, A.I., Loukidou, M.X. & Matis, K.A. (2004) Biosorption of
nucleotide sequence of pH11, an IncHI2 plasmid conferring toxic metals from aqueous solutions by bacteria strains isolated
multi-­antibiotic resistance and multi-­heavy metal resistance from metal-­polluted soils. Process Biochemistry, 39(8), 909–­916.
genes in a clinical Klebsiella pneumoniae isolate. Plasmid, 86, https://doi.org/10.1016/S0032​-9­ 592(03)00200​-­0
26–­31. https://doi.org/10.1016/j.plasm​id.2016.04.001
Zhang, M., Wan, K., Zeng, J., Lin, W., Ye, C. & Yu, X. (2020) Co-­
selection and stability of bacterial antibiotic resistance by arsenic How to cite this article: Vats, P, Kaur, UJ, Rishi, P
pollution accidents in source water. Environment International, (2022) Heavy metal-­induced selection and
135, 105351. https://doi.org/10.1016/j.envint.2019.105351 proliferation of antibiotic resistance: A review.
Zhang, S., Wang, Y., Song, H., Lu, J., Yuan, Z. & Guo, J. (2019) Copper
Journal of Applied Microbiology. 132:4058–4076.
nanoparticles and copper ions promote horizontal transfer of
plasmid-­mediated multi-­antibiotic resistance genes across bac-
https://doi.org/10.1111/jam.15492
terial genera. Environment International, 129, 478–­487. https://
doi.org/10.1016/j.envint.2019.05.054

You might also like