ArticlePDF AvailableLiterature Review

Abstract and Figures

Bacterial infections are raising serious concern across the globe. The effectiveness of conventional antibiotics is decreasing due to global emergence of multi-drug-resistant (MDR) bacterial pathogens. This process seems to be primarily caused by an indiscriminate and inappropriate use of antibiotics in non-infected patients and in the food industry. New classes of antibiotics with different actions against MDR pathogens need to be developed urgently. In this context, this review focuses on several ways and future directions to search for the next generation of safe and effective antibiotics compounds including antimicrobial peptides, phage therapy, phytochemicals, metalloantibiotics, lipopolysaccharide, and efflux pump inhibitors to control the infections caused by MDR pathogens.
This content is subject to copyright.
REVIEW ARTICLE
published: 13 May 2014
doi: 10.3389/fphar.2014.00105
Challenges and future prospects of antibiotic therapy: from
peptides to phages utilization
Santi M. Mandal 1*, Anupam Roy 1†, Ananta K. Ghosh1,Tapas K. Hazra2, Amit Basak1
and Octavio L. Franco3*
1Central Research Facility, Department of Chemistr y and Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
2Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston,TX, USA
3Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasilia, Brazil
Edited by:
Miguel Castanho, University of
Lisbon, Portugal
Reviewed by:
Frederico Nuno C. Aires Da Silva,
University of Lisbon, Portugal
Margarida Bastos, University of Porto,
Portugal
Marta Planas, University of Girona,
Spain
*Correspondence:
Santi M. Mandal, Central Research
Facility, Indian Institute ofTechnology
Kharagpur, Kharagpur - 721302, India
e-mail: mandalsm@gmail.com;
Octavio L. Franco, Centro de Análises
Proteômicas e Bioquímicas,
Pós-Graduação em Ciências
Genômicas e Biotecnologia,
Universidade Católica de Brasília,
SGAN 916N, Modulo C, Avenue W5,
Asa Norte, Sala 219, 70790-160
Brasilia, D. F., Brazil
e-mail: ocfranco@gmail.com
Santi M. Mandal and Anupam Roy
have contributed equally to this work.
Bacterial infections are raising serious concern across the globe. The effectiveness of
conventional antibiotics is decreasing due to global emergence of multi-drug-resistant
(MDR) bacterial pathogens.This process seems to be primarily caused by an indiscriminate
and inappropriate use of antibiotics in non-infected patients and in the food industry. New
classes of antibiotics with different actions against MDR pathogens need to be developed
urgently. In this context, this review focuses on several ways and future directions to search
for the next generation of safe and effective antibiotics compounds including antimicrobial
peptides, phage therapy, phytochemicals, metalloantibiotics, lipopolysaccharide, and efflux
pump inhibitors to control the infections caused by MDR pathogens.
Keywords: antibiotics, multi-drug-resistant pathogens, infection control
INTRODUCTION
Antibiotics are essential therapeutics, commonly used to con-
trol bacterial infections. They are one of the most significant
contributions to modern science and have proved to be of vital
importance in the dramatic rise in average life expectancy. Nev-
ertheless, antimicrobial resistance is clearly ready to jeopardize
this development now and in the near future. Four years after
the successful introduction of penicillin, the first appearance of
an antibiotic-resistant strain was reported during World War II
(Levy, 2002). Maurois (1959) warned about the deadly fact of
antibiotic resistance, stating that the inappropriate use of peni-
cillin could lead to the selection of resistant mutant forms” of
Staphylococcus aureus causing serious infections in the host. Since
then, acquired bacterial resistance has caused nosocomial infec-
tions with morbidity and mortality in hospitalized patients, and
to general alarm these infections have been observed spreading
to immune depressed patients (Michael et al., 2013). Each year
in the United States of America, at least two million persons
become infected with antibiotic-resistant bacteria and at least
23,000 people die every year as a direct result of such infec-
tions (Antibiotic resistance threats in the United States, 2013).
Many examples of resistant strains could be cited. Between 1987
and 2004, high levels of penicillin resistance in Streptococcus
pneumoniae were observed, reaching almost 20%. At the same
time, a 50% increase in methicillin-resistant in Staphylococcus
aureus (MRSA) was also observed (Herrmann and Laxminarayan,
2010). Additionally, very frequent and inappropriate use of antibi-
otics, lack of educational awareness and regulatory authority
regarding antibiotic usage, production, and marketing as well the
lack of infection control in hospitals and inadequate water and
sanitation in the community makes the situation worse. Spread of
Gram-negative bacilli resistance is an emerging problem of Asian
countries.
Surveillance study on the resistance on Salmonella enter-
ica serotype Typhi (Salmonella typhi) and Paratyphi (Salmonella
paratyphi) conducted in seven Asian countries (Korea, Taiwan,
Vietnam, Philippines, Singapore, Hong Kong, and Sri Lanka) from
2002 to 2004 emerged high rates of resistance against normally
used antibiotics. In Vietnam, the proportion of multi-drug-
resistant (MDR) strains was 30% higher than in the other six
countries (Chuang et al., 2008).
Currently, application of antibiotics seems to be the main
anti-infective solution for patients in major trauma or in inten-
sive care. Furthermore, similar antibiotic therapies are generally
applied to prevent post-surgery infections or in the treatment
of life-threatening infection in patients with various kinds of
www.frontiersin.org May 2014 |Volume 5 |Article 105 |1
Mandal et al. Challenges and future prospects of antibiotic therapy
cancer. These treatments, however, have become more difficult
due to pathogen resistance. Antibiotic resistance has led a string of
researchers to work on alternative strategies to“reset the clock” for
resistance levels in particular pathogens. Although some promis-
ing antibiotics have reached phase three trials, and many of
them are under phase two, the continuous development of new
compounds is extremely important, as will be described below.
In this context, this review article sheds some light on future
directions to search for the next generation of antimicrobial
compounds and examines strategies like antimicrobial peptides
(AMPs), phage therapy, phytochemicals, metallo-antibiotics,
lipopolysaccharide (LPS) inhibitors, and efflux pump inhibitors
to control the infections caused by MDR bacterial pathogens
(Table 1 ).
OVERVIEW OF MECHANISMS OF ANTIBIOTIC RESISTANCE
Antibiotic resistances are commonly relatedto bacterial mutations.
Such mutations could occur due to the selection pressure exerted
by the random and inappropriate use of bactericidal or bacte-
riostatic agents. Under continued selection pressure, the selected
bacteria may become resistant to antibiotics and spread to other
bacteria by transferring the resistance gene (Levy and Marshall,
2004). These unique resistance capabilities are generally subdi-
vided into four major issues. First is enzymatic drug inactivation,
as observed in the case of beta-lactamases (Davies, 1994). Sec-
ond, resistance could be related to alteration of specific target
sites (Spratt, 1994), as observed in the case of penicillin-binding
proteins (PBPs) in MRSA. Third, bacteria may acquire several
genes for a metabolic pathway. This alters bacterial cell walls and
thus makes antimicrobial agents incapable of binding to a bac-
terial target. Finally, the fourth issue is the reduction in drugs’
cellular uptake (Smith, 2004). In this case, para-amino benzoic
acid (PABA) is an important precursor for bacterial folic acid and
nucleic acid synthesis. Some sulphonamide-resistant bacteria do
not require PABA, instead using preformed folic acid as observed
in mammalian cells. As a result, a decrease in drug permeability
or an increase in active efflux of the drug across the cell surface
causes a decrease in drug accumulation in cellular compart-
ments (Nakaido, 1994). Bacteria may also acquire efflux pumps
that extrude the antibacterial agent from the cell before it can
reach its target site and exert its deleterious effect. This resistance
mechanism plays a vital role in reducing the clinical efficacy of
antibiotics. Moreover, the overproduction of efflux pumps is gen-
erally accompanied by a resistance improvement of two or more
Table 1 |Major types of antimicrobial compounds with their mechanisms of action.
Future therapy Mechanism Contemporary strategies to improve activity
Antimicrobial peptides Attach and insert into membrane bilayers to form
pores by “barrel-stave, “carpet, or
“toroidal-pore” mechanisms. DNA and
macromolecule synthesis inhibitors.
Optimization of peptide length and content of their sequences.
Conversion into peptidomimetics.
Generation of targeted antimicrobial peptides (Peptide antibiotic
conjugation).
Generation of antimicrobial peptides as prodrug candidates.
Antimicrobial peptides loaded into nanoparticle or micelles for sustained
release.
Phage therapy Bacteriophages are viruses that act as pathogens
against bacteria and completely lyse the bacteria.
Genetically engineered phages.
Genetically engineered phase as antibiotic delivery.
Engineered bacteriophage for phage targeted drug delivery.
Scale up of endolysin production.
Phytochemicals Multiple actions. Search for novel compounds and cost-effective methods of extraction
and purification of phytochemical.
Transgenic production in plant and microbial system to enhance number
of novel compounds.
Search for endophytic fungal metabolomics for the production of novel
compound of host.
Synthesis and modification of natural structure and analogs.
Metalloantibiotic Increased spectrum of conventional antibiotic
action.
Synthetic or semi-synthetic antimicrobial compound development
attaching metal to its structure.
In situ reducing and capping of metal nanoparticle with enhanced
antimicrobial activity.
Efflux pump inhibitor Molecules to inhibit the active protein pump in the
bacterial cell.
Chemical synthesis of effective efflux pumps inhibitor.
Screening of efflux pump inhibitors from natural origin and modifying
this compound synthetically.
Rationally designed transmembrane peptide mimics.
Frontiers in Pharmacology |Experimental Pharmacology and Drug Discovery May 2014 |Volume 5 |Article 105 |2
Mandal et al. Challenges and future prospects of antibiotic therapy
structurally unrelated antibiotics and significantly contributes to
the emergence of MDR pathogens. There are five major families
of efflux transporters, MFS (major facilitator superfamily), MATE
(multi-drug and toxic compound extrusion), RND (resistance
nodulation cell division) superfamily, SMR (small multi-drug-
resistance), and ABC (ATP-binding cassette) transporters (Eda
et al., 2011;Spellberg et al., 2013). All these mechanisms of resis-
tance have been targeted by the scientific community finding the
search for novel antibiotics with multiple functions, as described
above.
ANTIMICROBIAL PEPTIDES
Over the last few decades, several AMPs have been identified
(Figure 1) and rigorously investigated as alternatives to antibiotics.
They have been widely tested on the antibiotic-resistant bacte-
rial infections (Hancock and Lehrer, 1998;Ganz, 2003). AMPs
are the first line of defense in various organisms including plants
(Mandal et al., 2013;Roy et al., 2013), humans, insects and other
invertebrates, amphibians, birds, fish, and mammals (Martin et al.,
1995;Wang and Wang, 2004). Most AMPs are cationic in nature
and generally possess a specific amphipathic conformation. These
key players in defense systems have attracted extensive research
attention worldwide. AMPs are generally short (<100 amino acid
residues), positively charged and amphiphilic in nature. This
allows them to bind and insert themselves into membrane bilay-
ers to form pores by “barrel-stave, “carpet, or “toroidal-pore”
mechanisms (Matsuzaki et al., 1996;Oren and Shai, 1998;Yang
et al., 2001). Several data previously provided suggest that translo-
cate peptides may alter cytoplasmic membrane septum formation
(Salomon and Farias, 1992;Shi et al., 1996), inhibiting cell wall
synthesis (Brotz et al., 1998), bind to nucleic acids (Yonezawa et al.,
1992;Brotz et al., 1998;Park et al., 1998), inhibit nucleic acid syn-
thesis (Yonezawa et al., 1992;Boman et al., 1993;Silvestro et al.,
1997;Subbalakshmi and Sitaram, 1998), impede protein syn-
thesis (Yonezawa et al., 1992;Boman et al., 1993;Silvestro et al.,
1997;Subbalakshmi and Sitaram, 1998), or inhibit enzymatic
activity (Andreu and Rivas, 1998;Brogden, 2005). These fea-
tures make some AMPs most acceptable as a novel antibiotic class
and they can complement conventional antibiotic therapy (Mar-
shall and Arenas, 2003;Hancock and Sahl, 2006;Jenssen et al.,
2006).
The activity of AMPs may vary by switching amino acid com-
position, amphipathicity, cationic charges and size. In this context,
AMPs can be improved through the amalgamation of hydropho-
bic or charged amino acids, which has been revealed to modify
the selectivity for fungal and bacterial membranes (Davies, 1994;
Levy and Marshall, 2004). In this approach, different strategies in
designing novel peptides have been pursued, as described in sev-
eral studies (Tamamura et al., 1994;Pini et al., 2005). There are
descriptions of potential AMPs from natural sources helping to
design “tailor-made”AMPs owing to their easy availability through
solid-phase peptide synthesis (SPPS; Martin et al., 1995;Wang and
Wang, 2004). Several synthetic analogs of a number of naturally
occurring AMPs have been made, in an effort to identify signifi-
cant structural features that contribute to their enhanced activities
in vitro against Gram-positive and Gram-negative bacteria, fungi
as well as some enveloped viruses (Jenssen et al., 2006).
Cationic peptides contain cationic residues including arginines
and lysines. These residues are involved in the attraction of the neg-
atively charged bacterial cell surface. Structure–function studies of
FIGURE 1 |Some representative structure of antimicrobial peptides.
Structure represents beta defensin peptide from avian (AvBD2); an
amphipathic alpha-helical peptide from skin mucous of Pleuronectes
americanus (Pleurocidin); an antimicrobial hemolytic peptide from skin
of Bombina variegata (bombinin H2); skin secretion of Rana
temporaria (temporin); another antimicrobial peptide frog Xenopus
laevis (magainin) and peptide with helix-turn-helix motif from Rana
cascadae (ranatuerin).
www.frontiersin.org May 2014 |Volume 5 |Article 105 |3
Mandal et al. Challenges and future prospects of antibiotic therapy
host defense α-helical peptides have been carried out, with the aim
of designing diverse engineered cationic antimicrobial peptides
(eCAPs; Tencza et al., 1999;Jing et al., 2003;Phadke et al., 2003;
Deslouches et al., 2005a,b;Chan et al., 2006;Andrushchenko et al.,
2007,2008;Nguyen et al., 2010). Their focus was on structure–
function relationships of host-derived synthetic AMPs. A series
of eCAPs, called a lytic base unit (LBU) series, formed of only
Arg and Val, has been engineered to fold onto a flawless amphi-
pathic helical motifs in the occurrence of lipid membranes or
membrane mimitope solvents. Furthermore, Rozek et al. (2000)
explored the structure of the bovine AMP indolicidin linked
to dodecylphosphocholine and sodium dodecyl sulfate micelles.
Haney et al. (2012a) showed how specific amino acid side chains
influence the antimicrobial activity and structure of bovine lacto-
ferrampin. All the data have revealed the membrane perturbation
properties of Arg and Trp. Further research on the optimiza-
tion of the amphipathic helix has also been carried out. An
unusual series of eCAPs (6–18 residues long) has recently been
reported (Deslouches et al., 2013)tohavebroadandpotentin
vitro activity against MDR pathogens. These eCAPs consist exclu-
sively of Arg on the hydrophilic face and Trp on the hydrophobic
face.
Peptide modification, formulation, and delivery technologies
have also been explored to overcome the shortcomings of pharma-
cokinetics, bioavailability, and toxicity (Cole et al., 2003;Devocelle,
2012;Haney et al., 2012b). In this regard, several strategies have
been used, such as the optimization of peptide length and content,
offering an increase in selective antibacterial activity. Optimization
is generally done by minimizing the peptide length or switching
the peptide surface properties and systematically substituting each
residue. For these cases, computer-assisted AMP design is very
useful for an accurate estimation or prediction of the desired bio-
logical activity from the primary peptide structure (Fjell et al.,
2012). Moreover, conversion into peptidomimetics techniques is
able to improve the pharmacokinetic properties of AMPs, since
peptidomimetic structures are resistant to proteolysis.
Other strategies have also been applied to discovering novel
antimicrobials. Recently classical antibiotics were conjugated for
host defense peptide sequence, thereby increasing selectivity and
effectiveness against bacteria (Pokrovskaya and Baasov, 2010;
Arnusch et al., 2012). Additionally, the generation of AMP pro-
drug candidates has also been focused (Hancock, 2001;Stella,
2004;Gordon et al., 2005;Desgranges et al., 2012). Last but not
least, AMPs have been nanoencapsulated by strategies including
self-assembly, liposomes, polymeric structures, hydrogels, den-
dritic polymers, nanospheres, nanocapsules, carbon nanotubes,
and DNA cages. These strategies offer enhanced antimicrobial
activity, a reduction in collateral effects and also a clear protec-
tion from metabolic degradation (Urbán et al., 2012;Roy et al.,
2013).
The problem regarding the costs of synthesis and screening, sys-
temic, high manufacturing costs, and local toxicity, susceptibility
to proteolysis, sensitization, and allergic responses after repeated
uses of cationic membranolytic AMPs is the key barrier in success-
ful clinical application (Gordon et al., 2005). But AMPs, with their
unique multidirectional mode of action (Figure 2) and broad-
spectrum activities, rapid onset of killing, potentially low levels
FIGURE 2 |Schematic diagram of antibacterial peptides with
mechanism of action.
of induced resistance seem to represent one of the most promis-
ing future strategies to overcome increasing antibiotic-resistant
pathogens. These desirable and remarkable compounds are now
being studied extensively, and attempts to create them syntheti-
cally are being made by both academics and industries. Preclinical
and clinical studies of AMPs are being focused more in order to
overcome the problems. Efforts to produceAMPs on an industrial
large scale are now also in progress (Giuliani et al., 2007). A good
number of synthetic AMPs and at least 15 peptides or mimetics
are undergoing advanced clinical trials or have completed trials
as antimicrobial or immunomodulatory agents (Fjell et al., 2012).
Antimicrobial cationic lipopeptides like polymyxin, gramicidin S,
bracitracin, and cationic lantibiotic nisin have offered clinical effi-
cacy and are used widely (Laverty et al., 2010). In the future, the
inappropriate use of AMPs may lead to more resistant forms of
microorganisms that produce deadly infections. In this context,
innovative computer-assisted design strategies can strengthen the
ongoing development of next-generation therapeutic peptides and
peptide mimetics.
PHAGE THERAPY
The use of bacteriophages in controlling bacterial infections is
also a promising therapeutic option. Bacteriophages are bacterial
viruses that act as pathogens against bacteria. They show the abil-
ity of specifically attacking and killing only host bacterial cells at
the end of infection process (Sulakvelidze et al., 2001). After the
first isolation of bacteriophage in 1917, an oral phage prepara-
tion to treat bacterial dysentery was used (d’Herelle, 1917). Phages
are then extensively used and developed mainly in former Soviet
Union countries. Several commercial laboratories and compa-
nies in the USA, France and Germany developed phase products
(Hausler, 2006). The golden age in use of phage was in the 1930s,
but with antibiotics discovery, the progress of phage research and
use was reduced.
Bacteriophages have the ability to interfere between two cycles
lysogenic or lytic (Temperateness). In the lytic phage, the viral
DNA exists as a separate molecule within the bacterial cell, and
replicates separately from the host bacterial DNA. Each phage fol-
lows a unique pathway to control bacteria. Some of them show
a lytic infection cycle upon infecting their bacterial host. In this
Frontiers in Pharmacology |Experimental Pharmacology and Drug Discovery May 2014 |Volume 5 |Article 105 |4
Mandal et al. Challenges and future prospects of antibiotic therapy
case, they grow in high numbers in bacterial cells, leading to cel-
lular lyses. At the end of the cycle, a release of newly formed
phage particles is observed (Figure 3). Using the lysogenic path-
way, the phage genome integrates as part of the host genome. It
stays in a dormant state as a prophase for extended periods of
time. Adverse environmental conditions for the host bacterium
may activate the prophase, turning on the lytic cycle. At the end,
the newly formed phage particles are ready to lyse the host cell
(Skurnika and Strauch, 2006).
It has been noted that the bacterial mechanism of resistance
to phage seems to be lower when compared to antibiotics, which
are prone to bacterial resistance. The exponentially higher growth
kinetics generally overcomes bacterial growth (López et al., 1997).
Moreover, phages seem to show an extra advantage over com-
mon antibiotics, which are generally reduced by metabolism
and excretion, with several repeated administrations being neces-
sary (Inal, 2003). In the case of phages, increasing titers during
different periods removes the need for repeated doses. Addi-
tionally, high specificity for a particular bacterium does not
disturb the host-organism, so that phages do not affect com-
mensal intestine micro-flora, which is generally a side-effect in
the case of antibiotic ingestion (Inal, 2003). Although phages
may carry a virulence factor or toxic genes (Mesyanzhinov
et al., 2002;Brüssow et al., 2004;McGrath et al., 2004), a full
knowledge of phage genome sequences can address the possi-
ble complications during phage therapy (Skurnika and Strauch,
2006). Specific and non-toxic phages with high therapeutic
index can be applied to reduce the chances of opportunistic
pathogens.
Generally whole virulent phages are used as antibacterials.
Genetically modified phages are now also being studied, and have
been reported as useful in delivering antimicrobial agents to bac-
teria. Westwater et al. (2003) documented the use of a non-lytic
phage to precisely target and deliver DNA encoding bactericidal
proteins to target. Engineered bacteriophages can also enhance the
killing of antibiotic-resistant bacteria, persistent cells and biofilm
cells. They reduce the number of antibiotic-resistant bacteria
that ascend from an antibiotic-treated population, and act as a
robust adjuvant for other bactericidal antibiotics (Lu and Collins,
2009;Kaur et al., 2012). Moreover, a study on endolysins, which
are hydrolytic enzymes secreted by bacteriophage, has revealed
potential antimicrobial activity (Nelson et al., 2012).
FIGURE 3 |Mechanism of phage therapy. Image represents the schematic diagram of developmental cycle of lytic bacteriophage.
www.frontiersin.org May 2014 |Volume 5 |Article 105 |5
Mandal et al. Challenges and future prospects of antibiotic therapy
Treatment using the phage is not approved yet in countries
other than Russia and Georgia. Phages are currently being used
therapeutically only in the Russia and Georgia to treat extreme bac-
terial infections where conventional to antibiotics do not respond
(Karl, 2004;Parfitt, 2005). The use of phage technology for bacte-
rial control could also be applied in veterinary products focusing
on animal health. Bacterial resistance to antibiotics is a serious
concern for animal production, which could also be addressed by
phage therapy in the near future. Based on several features of bac-
teriophages, it has even been proposed that they be used in food
to prevent bacterial foodborne infections in food products and
on food contact surfaces (Borysowski et al., 2011). The example
includes LMP-102, which was regarded safe for use as food addi-
tive in meat and poultry products as an antibacterial agent against
Listeria monocytogenes (Daniells, 2006).
Archaebacteria-specific viruses or archeophages are the most
recent discoveries to show successful results in controlling bacte-
rial spread. Some bacteriophages have been used as anti-infective
agents, such as bacteriophage lysins and bacteriophage tail-like
bacteriocins (Schuch et al., 2002;Huff et al., 2004). For exam-
ple, a G phage lysine, PlyG, can effectively control Bacillus
anthraces in mice models (Schuch et al., 2002). Phages are recently
being commercialized in several areas of biological applica-
tions. Companies throughout the world like Intralytix (Baltimore,
MD, USA -product based on food safety), Phage Biotech Ltd
have (Rehovot, Israel -Anti-Pseudomonas infectives), BioCon-
trol (Southampton, UK -Pseudomonas infections of the ear),
EBI Food Safety (Wageningen, Netherlands -product based on
Food Safety. cocktail of phage against Listeria), JSC Biochim-
pharm (Tbilisi, Republic of Georgia -mixture of phage lysates),
Gangagen (India -phage against Staphylococcus aureus), Omni-
lytics (Salt Lake City, UT, USA -Agricultural use) etc., (Housby
and Mann, 2009). Despite the clear benefits of phage therapy,
some problems like development of antibodies after repeated
treatment with phages, rapid uptake and inactivation of phages
by spleen, contamination of therapeutic phage preparations
with endotoxins from bacterial debris, limited host range, reg-
ulation, bacterial resistance to phages, engineering, bacterial
lysis side effects and delivery should be scrutinized more thor-
oughly to make these potent therapeutics in the near future
(Inal, 2003;Lu and Koeris, 2011). In summary the phage
theraphy contribution in terms of continued investments in
research, development, and clinical trials from the public and
private sectors are needed to overcome regulatory and technical
hurdles.
PHYTOCHEMICALS
Phytochemicals are the tremendous gift of nature. They are the
secondary metabolites basically found in plants for specific func-
tional purposes. In most cases, these substances act as a plant
defense mechanism against microorganisms, insects and herbi-
vores. At the dawn of civilization, phytochemicals, in the form
of plants, were the only weapon in a struggle between man and
microbes. In recent times they have stimulated the same inter-
est both as fundamental sources of new chemical diversity and
integral components of today’s pharmaceutical compendium. But
with ever more incidents of MDR pathogens, the search for new
chemicals from plants offering a wide range of activity is the recent
focus of researchers.
At the moment thousands of compounds derived from plants
have been listed as antimicrobials. Phytochemicals within the
group of phenolics, terpenoids, essential oils, alkaloids, proteins
and peptides, possess potent antimicrobial potential with varying
mode of action (Cowan, 1999;Figure 4). Modern research is not
only confined to searching for antimicrobial compounds but has
also found several enzymatic inhibitors. Berberine is a hydropho-
bic cation found in common barberry (Berberis species) plants and
the medicinal plant goldenseal (Hydrastis canadensis). Although
the immutable targets and positive charge (facilitating active accu-
mulation in bacterial cells) makes berberine an efficient antibac-
terial, the fact that it is readily extruded by pathogen-encoded
MDR pumps rendered it ineffective. This limitation was overcome
by finding and applying another barberry-isolated compound,
5-methoxyhydnocarpin, which acts by blocking so-called major
facilitator MDRs of Gram-positive bacteria. In combination, the
two act as potent antimicrobials (Hsieh et al., 1998;Stermitz et al.,
2000;Lewis, 2001;Boucher et al., 2009). Furthermore, a newly
identified compound, 4-[N-(1,8-naphthalimide)]-n-butyric acid,
showed inhibition activity of the Vibrio cholerae transcriptional
regulator ToxT. Cholera toxin and the toxin-co-regulated pilus are
regulated by ToxT. This compound was tested in an animal model,
with infant mice, and was also reported to protect intestinal col-
onization by V. c h o l e r a (Hung et al., 2005;Lewis and Ausubel,
2006).
Strategies like study the combo effects of antibiotic and phyto-
chemical are recently been studied. Recently interaction studies are
drawn to study the associated effects of antibiotic and phytochemi-
cals (Sakharkar et al., 2009;Jayaraman et al., 2013). A current study
revealed that phytochemical-antibiotic conjugates have multitar-
get inhibitors of Pseudomononas aeruginosa GyrB/ParE and DHFR
was extremely effective (Jayaraman et al., 2010).
In the search for novel compounds and cost-effective methods
of extraction, purification of phytochemicals is a major concern.
FIGURE 4 |Representative chemical structures of some antimicrobial
phytochemicals. Phytochemicals like phenolics, saponins, essential oils,
terpenoids, alkaloids, and flavonids are the major classes showed
antibacterial activity.
Frontiers in Pharmacology |Experimental Pharmacology and Drug Discovery May 2014 |Volume 5 |Article 105 |6
Mandal et al. Challenges and future prospects of antibiotic therapy
But the mode of action derived from the structure may lead new
potent antibiotic and precursor molecule. For this, genetically
modified plant and microbial systems are also now being tried
to explore an enhanced number of novel compounds. Addition-
ally, novel antimicrobial structures have been created synthetically.
Synthetic analogs and modification of the structure may lead
to the discovery of novel structures with a broader spectrum of
activity.
METALLOANTIBIOTICS
Among different strategies to discover novel antibiotics,the incor-
poration of metal ions in antimicrobial compounds seems to be
promising. Metal ions perform an essential role in the functions
of synthetic and natural metalloantibiotics, being involved in very
specific interactions of such antibiotics with membranes, pro-
teins, nucleic acids, and several other biomolecules. This makes
metal ions effective in antibiotic structure or as an operative link-
age offering unique and specific bioactivities. Although several
antibiotics do not possess any metal ions in their structure, oth-
ers require metal ions for their proper functional activities. In
some cases metal ions are bound tightly to the antibiotic struc-
ture and regulate its action (Ming, 2003). Bacitracin, bleomycin,
streptonigrin, and albomycin are examples of such antibiotics.
In some cases metal ions are attached to the antibiotic molecule
without causing a major change in antibiotic structure. Tetra-
cyclines, aureolic acids, and quinolones are frequently used
in these strategies (Chohan et al., 2005). Synthetic or semi-
synthetic antimicrobial compounds also possess metal ions in their
structure.
In general, antimicrobials which contain metal compounds in
their structure in a natural form (nature-occurring) or which
have metal compounds incorporated synthetically are termed
metalloantibiotics. Transition metals are generally preferred in
metalloantibiotics and are present in very low concentration in
vivo. The ligand environment of transition metal ions can gener-
ally change considerably upon administration of a therapeutically
effective dose of an antibacterial drug (Sekhon, 2010). Some
strategies are followed to synthesize metal nanoparticles using
antibiotics as in situ reducing and capping agent. Here antibiotics
actasanin situ reducing and capping agent, thus offering potent
antimicrobial activities as well their application in antimicrobial
coatings (Jagannathan et al., 2007;Rai et al., 2010). Although
interactions with essential metal ions make more controllable
conditions for bacterial infections, the unwanted side effects like
toxicity and hemolytic activity sometimes increase simultaneously
(Heidenau et al., 2005). So the potential for oral administration or
internal use may be hampered.
EFFLUX PUMP INHIBITORS
One of the most important strategies to combat bacterial resis-
tance to antibiotics seems to be the efflux pump. Bacteria may
pump the drug out of the cell after its entrance, and among the
transporters involved in this pumping process are plasma mem-
brane translocases. Being non-specific in nature such transporters
are known as multi-drug-resistance pumps, being main determi-
nants of the antibiotic concentration inside a bacterial cell. Many
of them also act as drug/proton antiporters (protons enter the cell
as the drug leaves). This is a very common resistance mechanism
found in Escherichia coli, P. aeruginosa, Mycobacterium smegmatis,
and Staphylococcus aureus (Takiff et al., 1996). Depending upon
their varying structure and function, efflux pumps are subdivided
into five classes: SMR pumps of the drug/metabolite transporters
(DMTs) superfamily, ABC, RND, MFS, and MATE transporters
of the multi-drug/oligosaccharidyl-lipid/polysaccharide flippases
(MOP) superfamily (Piddock, 2006;Bhardwaj and Mohanty,
2012).
In order to restore the activity of antibiotics, an obvious
strategy consists of developing a compound that inhibits the
effects of efflux pump. Such molecules are named efflux pump
inhibitors (Figure 5). In order to get effective results, sev-
eral strategies have been taken including rational design of
efflux pump inhibitors, their chemical synthesis and potential-
ity as combination with commercial antibiotics (Marquez, 2005;
Delmar et al., 2014).
Although, there are few efflux pump inhibitors are available
in market and research in progress raises the hope that they
may be found in the near future. Screening of efflux pump
inhibitors from natural origins or under synthetic production
has attracted remarkable attention (Hudson et al., 2003). Struc-
tural modifications of such compounds may lead to an increase
in the spectrum of activities A wide number of effective chemical
compounds belonging to various chemical families have already
showed efflux pump inhibition (Brincat et al., 2011;Holler et al.,
2012a,b). Moreover, some plant-derived NorA EPIs and their
chemical modifications are carried out effectively (Thota et al.,
2008;Sabatini et al., 2011;Kalia et al., 2012). Recent rationally
designed transmembrane peptide mimics may work in efflux
pump inhibition. Maurya etal. (2013) have reported rationally
designed transmembrane peptide mimics of the multi-drug trans-
porter protein Cdr1. This acts as an antagonist to selectively
block drug efflux and chemosensitize azole-resistant Candida albi-
cans clinical isolates (Maurya et al., 2013). However, the major
advantages of efflux pump inhibitors are that the possibilities
on slower development of resistance by the target bacteria. Sev-
eral disadvantages are also documented including their chemical
synthesis due to bulky structure, solubility or permeability prob-
lems, required at higher concentration and chance of decreased the
activity at one or both target sites for steric or electronic config-
urations, unless these molecules are carefully designed (Bremner,
2007).
LPS INHIBITORS
The LPS layer or LPS in Gram-negative bacteria acts as a protec-
tive barrier. It prevents or slows down the entry of antibiotics
and another toxic compound that could kill or injure bacte-
ria (Miki and Hardt, 2013). LPS inhibitors are compounds that
generally work by inhibiting 3 deoxy-D-manno-octulosonic acid
8-phosphate synthase (KDO 8-p synthase), an important enzyme
in the LPS pathway. There have been several reports describ-
ing inhibitors, including PD404182 and polymyxin B (PMB),
which are compounds that have been applied with antibiotic
therapy, allowing the antibiotic to pass through the bacte-
rial cell wall (Palmer and Rifkind, 1974;Lindemann, 1988;
Birck et al., 2000).
www.frontiersin.org May 2014 |Volume 5 |Article 105 |7
Mandal et al. Challenges and future prospects of antibiotic therapy
FIGURE 5 |Few representative chemical structures of efflux pump inhibitors.
Among the strategies for exploiting LPS inhibitors more fully,
studies on bacterial enzymes that are essential for growth have
recently provoked much interest and will certainly attract more
attention in the near future in controlling bacterial resistance.
Some recently discovered molecules like pedopeptins are promis-
ing LPS inhibitor candidates, preventing bacterial growth (Cheung
et al., 2013;Kozuma et al., 2013). Some bacterial strains can
control other LPS functions, such as the ability of various Lac-
tobacillus strains to prevent Salmonella LPS-induced damage to
the epithelial barrier function (Yeung et al., 2013). This drug is
yet to be clinically validated with sufficient data. However, it is
likely that this and other small molecules of animal or plant ori-
gin or synthetically engineered may be found and developed soon
for use in inhibiting the translation machinery with an in vitro
system.
MYXOPYRONIN AND ARCHAEOCINS
Regulated gene expression is important in changing environ-
ments, stresses, and gene developmental programs. The activity
of transcriptional factors enables RNA polymerases (RNAPs)
to catalyze the transcription of DNA into RNA (Birck et al.,
2000;Wiesler et al., 2012). Myxopyronin is an α-pyrone antibi-
otic produced by Myxococcus fulvus offering broad-spectrum
antimicrobial activities for most Gram-positive species and some
Gram-negative bacteria [E. coli D21f2tolC (rfa tolC), Moraxella
catarrhalis ATCC25238]. It acts as inhibiting or binding bacterial
RNAP by changing the structure of the RNAP switch region of the
β-subunit of the enzyme. That renders the reading and transmit-
ting DNA code inactive, resulting in bacterial control (Irschik et al.,
1983;Campbell et al., 2001;Mukhopadhyay et al., 2008;Belogurov
et al., 2009;Ho et al., 2009;Srivastava et al., 2011). Rifampin, an
RNAP inhibitor in clinical utilization is capable of binds to the
β-subunit of RNAP within the DNA/RNA channel and blocks
the RNA elongation when the transcript converts two to three
nucleotides in length (Campbell et al., 2001). It is a broad spectrum
antimicrobial and is particularly active against M. tuberculosis.But,
the major problem in treatment failure and fatal clinical outcome
is due to the resistance to rifampin. The development of resistance
to rifampin is due to mutations in 81 base pair (27 codons) of the
β-subunit of RNAP (rpoB).
Archaea also contain potent antimicrobial compounds known
as archaeocins, which are archaeal proteinaceous antimicrobials.
Eight archaeocins from this family, among them halocins and
sulfolobicins, have been partially or fully characterized showing
antibacterial activity (O’Connor and Shand, 2002). The unique
mode of action offered by these groups of antimicrobials draws
remarkable attention as future antibiotic research. Besides, there
are lot of unknown bacterial and archaeal sources yet to be
explored. Thus discovery of new Myxopyronin and archaeocins
hinges on recovery and cultivation of bacterial and archaeal organ-
isms from the environment. Synthetic modification may lead this
compound to become a future potent drug.
CONCLUSION AND PROSPECTS
Growing concern about antibiotic resistance is propelling the
urgent modification of existing antibiotics and parallel devel-
opment of newer antibiotics. There are generally three inherent
pipelines available to fight against antibiotic resistance; i.e.,
antimicrobial chemical weaponry from natural products, syn-
thetic chemical compounds turned into antibiotics, and phages.
Antimicrobial compounds from natural product (AMPs, phyto-
chemicals, efflux pump inhibitors, LPS inhibitors myxopyronin,
Frontiers in Pharmacology |Experimental Pharmacology and Drug Discovery May 2014 |Volume 5 |Article 105 |8
Mandal et al. Challenges and future prospects of antibiotic therapy
and Archaeocins) have drawbacks regarding their isolation and
purification. The cost of production can be reduced by isolating
potent compounds from natural origins and then synthesizing
them, or by rationally modifying derived compounds. Phyto-
chemicals isolated from natural sources and then chemically
synthesized via modifications are likely to provide the most
effective antimicrobial drugs in the near future.
New compounds that target bacterial virulence can be devel-
oped to control the enormous threat posed by multi-drug-
resistance. Antibiotic structural modifications can be carried out
by synthesizing potent structures from already existing antibiotics.
Here the metalloantibiotics can play a great role.
In parallel, further research into toxicity against animal or
human cells, mechanisms of action, in vivo effects, and nega-
tive and positive interactions with common antibiotics should be
incorporated. The main challenge is to find the most effective
techniques for isolating and purifying newer and safer naturally
occurring antimicrobials against MDR pathogens. A better under-
standing of the structure, function and action mechanism of
existing and newly identified AMPs will lead to their being fine-
tuned by proper design to work against MDR pathogens. Phages
may also play a major role in treating bacterial infections in
humans. Combined treatment of phages with antibiotics is likely
to be a future choice. The problem regarding expansion of phages
can only be solved if large-scale clinical trials are carried out by
major pharmaceutical companies.
In summary, it is imperative to develop new classes of antibi-
otic or antimicrobial agents with different modes of action against
MDR pathogens. Combinational drug use is extensively used to
treat bacterial infection, but even this combinational dose pattern
may lead to resistance among pathogens. To overcome the chal-
lenges of antibiotic resistance, antimicrobial compounds with a
new mechanistic approach should be urgently sought.
REFERENCES
Andreu, D., and Rivas, L. (1998). Animal antimicrobial peptides: an overview.
Biopolymers 47, 415–433. doi: 10.1002/(SICI)1097-0282(1998)47:6<415::AID-
BIP2>3.0.CO;2-D
Andrushchenko, V. V., Aarabi, M. H., Nguyen, L. T., Prenner, E. J., and Vogel,
H. J. (2008). Thermodynamics of the interactions of tryptophan-rich cathelicidin
antimicrobial peptides with model and natural membranes. Biochim. Biophys.
Acta 1778, 1004–1014. doi: 10.1016/j.bbamem.2007.12.022
Andrushchenko, V. V., Vogel, H. J., and Prenner, E. J. (2007). Interactions of
tryptophan- rich cathelicidin antimicrobial peptides with model membranes
studied by differential scanning calorimetry. Biochim. Biophys. Acta 1768,
2447–2458. doi: 10.1016/j.bbamem.2007.05.015
Antibiotic resistance threats in the United States. (2013). Centers for Disease Control
and Prevention. Atlanta, GA: US Department of Health and Human Services.
Arnusch, C. J., Pieters, R. J., and Breukink, E. (2012). Enhanced membrane pore
formation through high- affinity targeted antimicrobial peptides. PLoS ONE
7:e39768. doi: 10.1371/journal.pone.0039768
Belogurov, G. A.,Vassylyeva, M. N., Sevostyanova, A., Appleman, J. R., Xiang, A. X.,
Lira, R., et al. (2009). Transcription inactivation through local refolding of the
RNA polymerase structure. Nature 457, 332–335. doi: 10.1038/nature07510
Bhardwaj, A. K., and Mohanty, P. (2012). Bacterial efflux pumps involved
in multidrug resistance and their inhibitors: rejuvinating the antimicro-
bial chemotherapy. Recent Pat. Antiinfect. Drug Disc. 7, 73–89. doi:
10.2174/157489112799829710
Birck, M. R., Holler, T. P., and Woodard,R. W. (2000). Identification of a slow tight-
binding inhibitor of 3-deoxy-d-manno-octulosonic acid 8-phosphate synthase.
J. Am. Chem. Soc. 122, 9334–9335. doi: 10.1021/ja002142z
Boman, H. G., Agerberth, B., and Boman, A. (1993). Mechanisms of action on
Escherichia coli of cecropin P1 and PR-39, two antibacterial peptides from pig
intestine. Infect. Immun. 61, 2978–2984.
Borysowski, J., Miêdzybrodzki, R., and Caister,A. G. (2011). Phage Therapy: Current
Research and Applications, eds J. Borysowski, R. Midezybrodzki, and A. Górski
(Poole: Caister Academic Press).
Boucher, H. W., Talbot, G. H., Bradley, J. S., Edwards, J. E., Gilbert, D., Rice, L. B.,
et al. (2009). Bad bugs, no drugs: no ESKAPE! An update from the Infectious
Diseases Society of America. Clin. Infect. Dis. 48, 1–12. doi: 10.1086/595011
Bremner, J. B. (2007). Some approaches to new antibacterial agents. Pure Appl.
Chem. 79, 2143–2153. doi: 10.1351/pac200779122143
Brincat, J. P., Carosati, E., Sabatini, S., Manfroni, G., Fravolini, A., Raygada, J. L.,
et al. (2011). Discovery of novel inhibitors of the NorA multidrug transporter of
Staphylococcus aureus.J. Med. Chem. 54, 354–365. doi: 10.1021/jm1011963
Brogden, K. A. (2005). Antimicrobial peptides: pore formers or metabolic inhibitors
in bacteria? Nat. Rev. Microbiol. 3, 238–250. doi: 10.1038/nrmicro1098
Brotz, H., Bierbaum, G., Leopold, K., Reynolds, P. E., and Sahl, H. G. (1998).
The lantibiotic mersacidin inhibits peptidoglycan synthesis by targeting lipid II.
Antimicrob. Agents Chemother. 42, 154–160. doi: 10.1186/1471-2180-8-186
Brüssow, H., Canchaya, C., and Hardt, W. D. (2004). Phages and the evolution
of bacterial pathogens: from genomic rearrangements to lysogenic conversion.
Microbiol. Mol. Biol. Rev. 68, 560–602. doi: 10.1128/MMBR.68.3.560-602.2004
Campbell, E. A., Korzheva, N., Mustaev, A., Murakami, K., Nair, S., Goldfarb, A.,
et al. (2001). Structural mechanism for rifampicin inhibition of bacterial rna
polymerase. Cell 104, 901–912. doi: 10.1016/S0092-8674(01)00286-0
Chan, D. I., Prenner, E. J., and Vogel, H. J. (2006). Tryptophan- and arginine-rich
antimicrobial peptides: structures and mechanisms of action. Biochim. Biophys.
Acta 1758, 1184–1202. doi: 10.1016/j.bbamem.2006.04.006
Cheung, S. T., Chang, D., Ming-Lum, A., and Mui, A. L. F. (2013). Interleukin-10
inhibits lipopolysaccharide induced miR- 155 precursor stability and maturation.
PLoS ONE 8:e71336. doi: 10.1371/journal.pone.0071336
Chohan, Z. H., Supuran, C. T., and Scozzafava, A. (2005). Metal binding and
antibacterial activity of ciprofloxacin complexes. J. Enzyme Inhib. Med. Chem. 20,
303–307. doi: 10.1080/14756360310001624948
Chuang, C. H., Su, L. H., Perera, J., Carlos, C., Tan, B. H., Kumarasinghe,
G., et al. (2008). Surveillance of antimicrobial resistance of Salmonella enter-
ica serotype Typhi in seven Asian countries. Epidemiol. Infect. 12, 1–4. doi:
10.1017/S0950268808000745
Cole, A. M., Liao, H. I., Ganz, T., and Yang, O. O. (2003). Antibacterial activity of
peptides derived from envelope glycoproteins of HIV-1. FEBS Lett. 535, 195–199.
doi: 10.1016/S0014-5793(02)03860-7
Cowan, M. M. (1999). Plant products as antimicrobial agents. Clin. Microbiol. Rev.
12, 564–582.
d’Herelle, F. (1917). Sur un microbe invisible antagoniste des bacteries dysen-
teriques. C. R. Acad. Sci. 165, 373–375.
Daniells, S. (2006). FDA Approves Viruses as Food Additive for Meats. Available at:
http://www.foodnavigator-usa.com/Suppliers2/FDA-approves-viruses-as-food-
additive-for-meats
Davies, J. (1994). Inactivation of antibiotics and the dissemination of resistance
genes. Science 15, 375–382. doi: 10.1126/science.8153624
Delmar, J.A., Su, C. C., and Yu, E.W. (2014). Bacterial multidrug efflux transpor ters.
Annu. Rev. Biophys. doi: 10.1146/annurev-biophys-051013-022855 [Epub ahead
of print].
Desgranges, S., Ruddle, C. C., Burke, L. P., McFadden, T. M., O’Brien, J. E.,
Fitzgerald-Hughes, D., et al. (2012). β-Lactam-host defense peptide conjugates as
antibiotic prodrug candidates targeting resistant bacteria. RSC Adv. 2, 2480–2492.
doi: 10.1039/c2ra01351g
Deslouches, B., Islam, K., Craigo, J. K., Paranjape, S. M., Montelaro, R. C.,
and Mietzner, T. A. (2005a). Activity of the de novo engineered antimicrobial
peptide WLBU2 against Pseudomonas aeruginosa in human serum and whole
blood: implications for systemic applications. Antimicrob. Agents Chemother. 49,
3208–3216. doi: 10.1128/AAC.49.8.3208-3216.2005
Deslouches, B., Phadke, S. M., Lazarevic, V., Cascio, M., Islam, K., Montelaro, R. C.,
et al. (2005b). De novo generation of cationic antimicrobial peptides: influence of
length and tryptophan substitution on antimicrobial activity. Antimicrob. Agents
Chemother. 49, 316–322. doi: 10.1128/AAC.49.1.316-322.2005
Deslouches, B., Steckbeck, J. D., Craigo, J. K., Doi, Y., Mietzner, T. A., and Monte-
laro, R. C. (2013). Rational design of engineered cationic antimicrobial peptides
www.frontiersin.org May 2014 |Volume 5 |Article 105 |9
Mandal et al. Challenges and future prospects of antibiotic therapy
consisting exclusively of arginine and tryptophan, and their activity against
multidrug-resistant pathogens. Antimicrob. Agents. Chemother. 57, 2511–2521.
doi: 10.1128/AAC.02218-12
Devocelle, M. (2012). Targeted antimicrobial peptides. Front. Immunol. 3:309. doi:
10.3389/fimmu.2012.00309
Eda, S., Mitsui, H., and Minamisawa, H. (2011). Involvement of the SmeAB mul-
tidrug efflux pump in resistance to plant antimicrobials and contribution to
nodulation competitiveness in Sinorhizobium meliloti.Appl. Environ. Microbiol.
77, 2855–2862. doi: 10.1128/AEM.02858-10
Fjell, C. D., Hiss, J. A., Hancock, R. E. W., and Schneider, G. (2012). Designing
antimicrobial peptides: form follows function. Nat. Rev. Drug Discov. 11, 37–51.
doi: 10.1038/nrd3591
Ganz, T. (2003). Defensins: antimicrobial peptides of innate immunity. Nat. Rev.
Immunol. 3, 710–720. doi: 10.1038/nri1180
Giuliani, A., Pirri, G., and Nicoletto, S. F. (2007). Antimicrobial peptides: an
overview of a promising class of therapeutics. CEJB 2, 1–33. doi: 10.2478/s11535-
007-0010-5
Gordon, Y. J., Romanowski, E. G., and McDermott, A. M. (2005). A review of
antimicrobial peptides and their therapeutic potential as anti infective drugs.
Curr. Eye Res. 30, 505–515. doi: 10.1080/02713680590968637
Hancock, R. E., and Lehrer, R. (1998). Cationic peptides: a new source of antibiotics.
Trends Biotechnol. 16,82–88. doi: 10.1016/S0167-7799(97)01156-6
Hancock, R. E. W. (2001). Cationic peptides: effectors in innate immunity
and novel antimicrobials. Lancet Infect. Dis. 1, 156–164. doi: 10.1016/S1473-
3099(01)00092-5
Hancock, R. E. W., and Sahl, H. G. (2006). Antimicrobial and host-defense peptides
as new anti-infective therapeutic strategies. Nat. Biotechnol. 24, 1551–1557. doi:
10.1038/nbt1267
Haney, E. F., Nazmi, K., Bolscher, J. G., and Vogel, H. J. (2012a). Influence of specific
amino acid side chains on the antimicrobial activity and structure of bovine
lactoferrampin. Biochem. Cell Biol. 90, 362–377. doi: 10.1139/o11-057
Haney, E. F., Nguyen, L. T., Schibli, D. J., and Vogel, H. J. (2012b). Design of a novel
tryptophan-rich membrane-active antimicrobial peptide from the membrane-
proximal region of the HIV glycoprotein, gp41. Beilstein J. Org. Chem. 8, 1172–
1184. doi: 10.3762/bjoc.8.130
Hausler, T. (2006). Viruses vs. Superbugs. A Solution to the Antibiotic Crisis? New
York: Macmillan.
Heidenau, F., Mittelmeier, W., Detsch, R., Haenle, M., Stenzel, F., Ziegler, G., et al.
(2005). A novel antibacterial titania coating: metal ion toxicity and in vitro surface
colonization. J. Mater. Sci. Mater. Med. 16, 883–888. doi: 10.1007/s10856-005-
4422-3
Herrmann, M., and Laxminarayan, R. (2010). Antibiotic effectiveness: new chal-
lenges in natural resource management. Annu.Rev.Resour.Econ. 2, 125–138. doi:
10.1146/annurev.resource.050708.144125
Ho, M. X., Hudson, B. P., Das, K., Arnold, E., and Ebright, R. H. (2009). Structures
of RNA polymerase-antibiotic complexes. Curr. Opin. Struct. Biol. 19, 715–723.
doi: 10.1016/j.sbi.2009.10.010
Holler, J. G., Christensen, S. B., Slotved, H. C., Rasmussen, H. B., Guzman,
A., Olsen, C. E., et al. (2012a). Novel inhibitory activity of the Staphylo-
coccus aureus NorA efflux pump by a kaempferol rhamnoside isolated from
Persea lingue Nees. J. Antimicrob. Chemother. 67, 1138–1144. doi: 10.1093/jac/
dks005
Holler,J. G., Slotved, H. C., Molgaard, P., Olsen, C. E., and Christensen, S. B. (2012b).
Chalcone inhibitors of the NorA efflux pump in Staphylococcus aureus whole cells
and enriched everted membrane vesicles. Bioorg. Med. Chem. 20, 4514–4521. doi:
10.1016/j.bmc.2012.05.025
Housby, J. N., and Mann, N. H. (2009). Phage therapy. Drug Discov. Today 14,
536–540. doi: 10.1016/j.drudis.2009.03.006
Hsieh, P. C., Siegel, S. A., Rogers, B., Davis, D., and Lewis, K. (1998). Bacteria lacking
a multidrug pump: a sensitive tool for drug discovery. Proc. Natl. Acad. Sci. U.S.A.
95, 6602–6606. doi: 10.1073/pnas.95.12.6602
Hudson, A., Imamura, T., Gutteridge, W., Kanyok, T., and Nunn, P. (2003).
The Current Anti-TB Drug Research and Development Pipeline. Available at:
http://www.who.int/tdr/publications/documents/anti-tb-drug.pdf
Huff, W. E., Huff, G. R., Rath, N. C., Balog, J. M., and Donoghue, A. M. (2004).
Therapeutic efficacy of bacteriophage and Baytril (enrofloxacin) individually and
in combination to treat colibacillosis in broilers. Poult. Sci. 83, 1944–1947. doi:
10.1093/ps/83.12.1944
Hung, D. T., Shakhnovich, E. A., Pierson, E., and Mekalanos, J. J. (2005). Small-
molecule inhibitor of Vibrio cholerae virulence and intestinal colonization. Science
310, 670–674. doi: 10.1126/science.1116739
Inal, J. M. (2003). Phage therapy: a reappraisal of bacteriophages as antibiotics.
Arch. Immunol. Ther. Exp. (War sz.) 51, 237–244.
Irschik, H., Gerth, K., Höfle, G., Kohl, W., and Reichenbach, H. (1983). The
myxopyronins, new inhibitors of bacterial RNA synthesis from Myxococcus
fulvus (Myxobacterales). J. Antibiot.(Tokyo) 36, 1651–1658. doi: 10.7164/antibi-
otics.36.1651
Jagannathan, R., Poddar, P., and Prabhune, A. (2007). Cephalexin-mediated synthe-
sis of quasi-spherical and anisotropic gold nanoparticles and their in situ capping
by the antibiotic. J. Phys. Chem. C 111, 6933–6938. doi: 10.1021/jp067645r
Jayaraman, P., Sakharkar, K. R., Lim, C., Siddiqi, M. I., Dhillon, S. K., and Sakharkar,
M. K. (2013). Novel phytochemical-antibiotic conjugatesas multitarget inhibitors
of Pseudomononas aeruginosa GyrB/ParE and DHFR. Drug Des. Devel. Ther.7,
449–475. doi: 10.2147/DDDT.S43964
Jayaraman, P., Sakharkar, M. K., Lim, C. S., Tang, T. H., and Sakharkar, K.
R. (2010). Activity and interactions of antibiotic and phytochemical combina-
tions against Pseudomonas aeruginosa in vitro. Int. J. Biol. Sci. 6, 556–568. doi:
10.7150/ijbs.6.556
Jenssen, H., Hamill, P., and Hancock, R. E. W. (2006). Peptide antimicrobial agents.
Clin. Microbiol. Rev. 19, 491–511. doi: 10.1128/CMR.00056-05
Jing, W., Hunter, H. N., Hagel, J., and Vogel, H. J. (2003). The structure of the
antimicrobial peptide Ac-RRWWRF-NH2 bound to micelles and its interactions
with phospholipid bilayers. J. Peptide Res. 61, 219–229. doi: 10.1034/j.1399-
3011.2003.00050.x
Kalia, N. P., Mahajan, P., Mehra,R., Nargotra, A., Sharma, J. P.,Koul, S., et al. (2012).
Capsaicin, a novel inhibitor of the NorA efflux pump, reduces the intracellular
invasion of Staphylococcus aureus.J. Antimicrob. Chemother. 67, 2401–2408. doi:
10.1093/jac/dks232
Karl, T. (2004). Old dogma, new tricks–21st Century phage therapy. Nat. Biotechnol.
22, 31–36. doi: 10.1038/nbt0104-31
Kaur, T., Nafissi, N., Wasfi, O., Sheldon, K., Wettig, S., and Slavcev, R. (2012).
Immunocompatibility of bacteriophages as nanomedicines. J. Nanotech. 2012,
1–13. doi: 10.1155/2012/247427
Kozuma, S., Takahata, Y. H., Fukuda, D., Kuraya, N., Nakajima, M., and Ando, O.
(2013). Screening and biological activities of pedopeptins, novel inhibitors of LPS
produced by soil bacteria. J. Antibiot. 67, 237–242.
Laverty, G., McLaughlin, M., Shaw, C., Gorman, S. P., and Gilmore, B. F. (2010).
Antimicrobial activity of short, synthetic cationic lipopeptides. Chem. Biol. Drug
Des. 75, 563–569. doi: 10.1111/j.1747-0285.2010.00973.x
Levy, S. B. (2002). The Antibiotic Paradox: How the Misuse of Antibiotics Destroys
their Curative Powers, 2nd Edn. Cambridge, MA: Perseus Publishing.
Levy, S. B., and Marshall, B. (2004). Antibacterial resistance worldwide: causes,
challenges and responses. Nat. Med. 10, S122–S129. doi: 10.1038/nm1145
Lewis, K. (2001). In search of natural substrates and inhibitors of MDR pumps.
J. Mol. Microbiol. Biotechnol. 3, 247–254.
Lewis, K., and Ausubel,F. M. (2006). Prospects for plant-derived antibacterials. Nat.
Biotechnol. 24, 1504–1507. doi: 10.1038/nbt1206-1504
Lindemann, R. A. (1988). Bacterial activation of human natural killer cells: role of
cell surface lipopolysaccharide. Infect. Immun. 56, 1301–1308.
López, R., García, E., García, P., and García, J. L. (1997). The pneumococcal cell wall
degrading enzymes: a modular design to create new lysins? Microb. Drug Resist.
3, 199–211. doi: 10.1089/mdr.1997.3.199
Lu, T. K., and Collins, J. J.(2009). Eng ineeredbacter iophage targeting gene networks
as adjuvants for antibiotic therapy. Proc. Natl. Acad. Sci. U.S.A. 106, 4629–4634.
doi: 10.1073/pnas.0800442106
Lu, T. K., and Koeris, M. S. (2011). The next generation of bacteriophage therapy.
Curr. Opin. Microbiol. 14, 524–531. doi: 10.1016/j.mib.2011.07.028
Mandal, S. M., Sharma, S., Pinnaka, A. K., Kumari, A., and Korpole, S. (2013).
Isolation and characterization of diverse antimicrobial lipopeptides produced by
Citrobacter and Enterobacter.BMC Microbiol. 13:152. doi: 10.1186/1471-2180-
13-152
Marquez, B. (2005). Bacterial efflux systems and efflux pumps inhibitors. Biochimie
87, 1137–1147. doi: 10.1016/j.biochi.2005.04.012
Marshall, S. H., and Arenas, G. (2003). Antimicrobial peptides: a natural alternative
to chemical antibiotics and a potential for applied biotechnology. Electron. J.
Biotechnol. 6:2. doi: 10.2225/vol6-issue3-fulltext-1
Frontiers in Pharmacology |Experimental Pharmacology and Drug Discovery May 2014 |Volume 5 |Article 105 |10
Mandal et al. Challenges and future prospects of antibiotic therapy
Martin, E., Ganz, T., and Lehrer, R. I. (1995). Defensins and other endogenous
peptide antibiotics of vertebrates. J. Leukoc. Biol. 58, 128–136.
Matsuzaki, K., Murase, O., Fujii, N., and Miyajima, K. (1996). An antimicro-
bial peptide, magainin 2, induced rapid flip-flop of phospholipids coupled with
pore formation and peptide translocation. Biochemistry 35, 11361–11368. doi:
10.1021/bi960016v
Maurois, A. (1959). Life of Sir Alexander Fleming: Discoverer of Penicillin. London:
E. P. Dutton & Co., Inc.
Maurya, I. K., Thota, C. K., Verma, S. D., Sharma, J., Rawal, M. K., Ravikumar,
B., et al. (2013). Rationally designed transmembrane peptide mimics of the mul-
tidrug transporter protein Cdr1 act as antagonists to selectively block drug efflux
and chemosensitize azole-resistant clinical isolates of Candida albicans.J. Biol.
Chem. 288, 16775–16787. doi: 10.1074/jbc.M113.467159
McGrath, S., Fitzgerald, G. F., and van Sinderen, D. (2004). The impact
of bacteriophage genomics. Curr. Opin. Biotechnol. 15, 94–99. doi:
10.1016/j.copbio.2004.01.007
Mesyanzhinov, V. V., Robben, J., Grymonprez, B., Kostyuchenko, V. A., Bourkalt-
seva, M. V., Sykilinda, N. N., et al. (2002). The genome of bacteriophage fKZ of
Pseudomonas aeruginosa.J. Mol. Biol. 317, 1–19. doi: 10.1006/jmbi.2001.5396
Michael, P. D., Loneragan, G. H., Scott, M., and Singer, R. S. (2013). Antimicrobial
resistance: challenges and perspectives. Compr. Rev. Food Sci. Food Saf. 12, 234–
248. doi: 10.1111/1541-4337.12008
Miki, T., and Hardt, W.-D. (2013). Outer membrane permeabilization is an essential
step in the killing of gram-negative bacteria by the lectin RegIIIβ.PLoS ONE
8:e69901. doi: 10.1371/journal.pone.0069901
Ming, L. J. (2003). Structure and function of “metalloantibiotics.” Med. Res. Rev. 23,
697–762. doi: 10.1002/med.10052
Mukhopadhyay, J., Das, K., Ismail, S., Koppstein, D., Jang, M., Hudson, B., et al.
(2008). The RNA polymerase “switch region is a target for inhibitors. Cell 135,
295–307. doi: 10.1016/j.cell.2008.09.033
Nakaido, H. (1994). Prevention of drug access to bacterial targets: permeability
barriers and active efflux. Science 15, 382–388. doi: 10.1126/science.8153625
Nelson, D. C., Schmelcher, M., Rodriguez-Rubio, L., Klumpp, J., Pritchard, D. G.,
Dong, S., et al. (2012). Endolysins as antimicrobials. Adv. Virus Res. 83, 299–365.
doi: 10.1016/B978-0-12-394438-2.00007-4
Nguyen, L. T., Chau, J. K., Perry, N. A., de Boer, L., Zaat, S. A., and Vogel, H. J.
(2010). Serum stabilities of short tryptophan- and arginine-rich antimicrobial
peptide analogs. PLoS ONE 5:12684. doi: 10.1371/journal.pone.0012684
O’Connor,E. M., and Shand,R. F. (2002). Halocins and sulfolobicins: the emerging
story of archaeal protein and peptide antibiotics. J. Ind. Microbiol. Biotechnol. 28,
23–31. doi: 10.1038/sj/jim/7000190
Oren, Z., and Shai, Y. (1998). Mode of action of linear amphipathic α-helical
antimicrobial peptides. Biopolymers 47, 451–463. doi: 10.1002/(SICI)1097-
0282(1998)47:6<451::AID-BIP4>3.0.CO;2-F
Palmer,J. D., and Rifkind, D. (1974). Neutralization of the hemodynamic effects of
endotoxin by polymyxin B. Surg. Gynecol. Obstet. 138, 755–759.
Parfitt, T. (2005). Georgia: an unlikely stronghold for bacteriophage therapy. Lancet
365, 2166–2167. doi: 10.1016/S0140-6736(05)66759-1
Park, C. B., Kim, H. S., and Kim, S. C. (1998). Mechanism of action of the antimi-
crobial peptide buforin II: buforin II kills microorganisms by penetrating the cell
membrane and inhibiting cellular functions. Biochem. Biophys. Res. Commun.
244, 253–257. doi: 10.1006/bbrc.1998.8159
Phadke, S. M., Islam, K., Deslouches, B., Kapoor, S. A., Beer Stolz, D., Watkins, S.
C., et al. (2003). Selective toxicity of engineered lentivirus lytic peptides in a CF
airway cell model. Peptides 24, 1099–1107. doi: 10.1016/j.peptides.2003.07.001
Piddock, L. J. (2006). Clinically relevant chromosomally encoded multidrug
resistance efflux pumps in bacteria. Clin. Microbiol. Rev. 19, 382–402. doi:
10.1128/CMR.19.2.382-402.2006
Pini, A., Giuliani, A., Falciani, C., Runci,Y., Ricci, C., Lelli, B., et al. (2005). Antimi-
crobial activity of novel dendrimeric peptides obtained by phage display selection
and rational modification. Antimicrob. Agents Chemother. 49, 2665–2672. doi:
10.1128/AAC.49.7.2665-2672.2005
Pokrovskaya,V., and Baasov, T. (2010). Dual-acting hybrid antibiotics: a promising
strategy to combat bacterial resistance. Expert Opin. Drug Discov. 5, 883–902. doi:
10.1517/17460441.2010.508069
Rai, A., Prabhune, A., and Perry, C. C. (2010). Antibiotic mediated synthesis of
gold nanoparticles with potent antimicrobial activity and their application in
antimicrobial coatings. J. Mater. Chem. 20, 6789–6798. doi: 10.1039/c0jm00817f
Roy, A., Franco, O. L., and Mandal, S. M. (2013). Biomedical exploitation of self-
assembled peptide based nanostructures. Curr. Protein Pept. Sci. 14, 580–587. doi:
10.2174/1389203711209070687
Rozek, A., Friedrich, C. L., and Hancock, R. E. W. (2000). Structure of the
bovine antimicrobial peptide indolicidin bound to dodecylphosphocholine and
sodium dodecyl sulfate micelles. Biochemistry 39, 15765–15774. doi: 10.1021/
bi000714m
Sabatini, S., Gosetto, F., Manfroni, G., Tabarrini, O., Kaatz, G. W., Patel,
D., et al. (2011). Evolution from a natural flavones nucleus to obtain 2-(4-
Propoxyphenyl)quinoline derivatives as potent inhibitors of the S. aureus NorA
efflux pump. J. Med. Chem. 54, 5722–5736. doi: 10.1021/jm200370y
Sakharkar, M. K., Jayaraman, P., Soe, W. M., Chow, V. T., Sing, L. C., and Sakharkar,
K. R. (2009). In vitro combinations of antibiotics and phytochemicals against
Pseudomonas aeruginosa.J. Microbiol. Immunol. Infect. 42, 364–370.
Salomon, R. A., and Farias, R. N. (1992). Microcin 25,a novel antimicrobial peptide
produced by Escherichia coli.J. Bacteriol. 174, 7428–7435.
Schuch, R., Nelson, D., and Fischetti,V. A. (2002). A bacteriolytic agent that detects
and kills Bacillus anthracis.Nature 418, 884–889. doi: 10.1038/nature01026
Sekhon, B. S. (2010). Metalloantibiotics and antibiotic mimics an overview.
J. Pharm. Educ. Res. 1, 1–20.
Shi, J., Ross, C. R., Chengappa, M. M., Sylte, M. J., McVey, D. S., and Blecha, F.
(1996). Antibacterial activity of a synthetic peptide (PR-26) derived from PR-
39, a proline-arginine-rich neutrophil antimicrobial peptide. Antimicrob. Agents
Chemother. 40, 115–121.
Silvestro, L., Gupta, K., Weiser, J. N., and Axelsen, P. H. (1997). The concentration-
dependent membrane activity of cecropin A. Biochemistry 36, 11452–11460. doi:
10.1021/bi9630826
Skurnika, M., and Strauch, E. (2006). Phage therapy: facts and fiction. Int. J. Med.
Microbiol. 296, 5–14. doi: 10.1016/j.ijmm.2005.09.002
Smith, A. (2004).“Bacterial resistance to antibiotics,”in Hugo and Russell’s Pharma-
ceutical Microbiology, 7th Edn, eds S. P. Denyer, N. A. Hodges, and S. P. Gorman
(Malden, MA: Blackwell Science).
Spellberg, B., Bartlett, J. G., and Gilbert, D. N. (2013). The future of antibiotics and
resistance. N.Engl.J.Med.368, 299–302. doi: 10.1056/NEJMp1215093
Spratt, B.G. (1994). Resistance to antibiotics mediated by target alterations. Science
15, 388–393. doi: 10.1126/science.8153626
Srivastava, A., Talaue, M., Liu, S., Degen, D., Ebright, R. Y., Sineva, E., etal. (2011).
New target for inhibition of bacterial RNA polymerase: “switch region.” Curr.
Opin. Microbiol. 14, 532–543. doi: 10.1016/j.mib.2011.07.030
Stella, V. J. (2004). Prodrugs as therapeutics. Expert Opin. Ther. Patents 14, 277–280.
doi: 10.1517/13543776.14.3.277
Stermitz, F. R., Lorenz, P., Tawara, J. N., Zenewicz, L. A., and Lewis, K. (2000).
Synergy in a medicinal plant: antimicrobial action of berberine potentiated by 5’-
methoxyhydnocarpin, a multidrug pump inhibitor. Proc. Natl. Acad. Sci. U.S.A.
97, 1433–1437. doi: 10.1073/pnas.030540597
Subbalakshmi, C., and Sitaram, N. (1998). Mechanism of antimicrobial action
of indolicidin. FEMS Microbiol. Lett. 160, 91–96. doi: 10.1111/j.1574-
6968.1998.tb12896.x
Sulakvelidze, A., Alavidze, Z., and Morris, J. G. Jr. (2001). Bacteriophage Therapy.
Antimicrob. Agents Chemother. 45, 649–659. doi: 10.1128/AAC.45.3.649-659.2001
Takiff, H. E., Cimino, M., Musso, M. C., Weisbrod, T., Martinez, R., Delgado, M.
B., et al. (1996). Efflux pump of the proton antiporter family confers low-level
fluoroquinolone resistance in Mycobacterium smegmatis.Proc. Natl. Acad. Sci.
U.S.A. 93, 362–366. doi: 10.1073/pnas.93.1.362
Tamamura, H., Murakami, T., Masuda, M., Otaka, A., Takada, W., Ibuka, T.,
et al. (1994). Structure- activity relationships of an anti-HIV peptide, T22.
Biochem. Biophys. Res. Commun. 205, 1729–1735. doi: 10.1006/bbrc.1994.
2868
Tencza, S. B., Creighton, D. J., Yuan, T., Vogel, H. J., Montelaro, R. C., and Mietzner,
T. A. (1999). Lentivirus-derived antimicrobial peptides: increased potency by
sequence engineering and dimerization. J. Antimicrob. Chemother. 44, 33–41.
doi: 10.1093/jac/44.1.33
Thota, N., Koul, S., Reddy, M. V., Sangwan, P. L., Khan, I. A., Kumar,A., et al. (2008).
Citral derived amides as potent bacterial NorA efflux pump inhibitors. Bioorg.
Med. Chem. 16, 6535–6543. doi: 10.1016/j.bmc.2008.05.030
Urbán, P., Valle-Delgado, J. J., Moles, E., Marques, J., Díez, C., and Fernàndez-
Busquets, X. (2012). Nanotools for the delivery of antimicrobial peptides. Curr.
Drug Targets 13, 1158–1172. doi: 10.2174/138945012802002302
www.frontiersin.org May 2014 |Volume 5 |Article 105 |11
Mandal et al. Challenges and future prospects of antibiotic therapy
Wang, Z., and Wang, G. (2004). APD: the antimicrobial peptide database. Nucleic
Acids Res. 32, 590–592. doi: 10.1093/nar/gkh025
Westwater, C., Kasman, L. M., Schofield, D. A., Werner, P. A., Dolan, J.
W., Schmidt, M. G., et al. (2003). Use of genetically engineered phage to
deliver antimicrobial agents to bacteria: an alternative therapy for treatment
of bacterial infections. Antimicrob. Agents Chemother. 47, 1301–1307. doi:
10.1128/AAC.47.4.1301-1307.2003
Wiesler, S. C., Burrows, P. C., and Buck, M. (2012). A dual switch controls bacte-
rial enhancer-dependent transcription. Nucleic Acids Res. 40, 10878–10892. doi:
10.1093/nar/gks844
Yang, L., Harroun, T. A., Weiss, T. M., Ding, L., and Huang, H. W. (2001). Barrel-
stave model or toroidal model? A case study on melittin pores. Biophys. J. 81,
1475–1485. doi: 10.1016/S0006-3495(01)75802-X
Yeung, C. Y., Chiang Chiau, J. S., Chan, W. T., Jiang, C. B., Cheng, M. L., Liu, H. L.,
et al. (2013). In vitro prevention of Salmonella lipopolysaccharide-induced dam-
ages in epithelial barrier function by various Lactobacillus strains. Gastroenterol.
Res. Pract. 2013, 6. doi: 10.1155/2013/973209
Yonezawa, A., Kuwahara, J., Fujii, N., and Sugiura, Y. (1992). Binding of tachyplesin
I to DNA revealed by footprinting analysis: significant contribution of secondary
structure to DNA binding and implication for biological action. Biochemistry 31,
2998–3004. doi: 10.1021/bi00126a022
Conflict of Interest Statement: The authors declare that the research was conducted
in the absence of any commercial or financial relationships that could be construed
as a potential conflict of interest.
Received: 10 March 2014; accepted: 22 April 2014; published online: 13 May 2014.
Citation: Mandal SM, Roy A, Ghosh AK, Hazra TK, Basak A and Franco OL
(2014) Challenges and future prospects of antibiotic therapy: from peptides to phages
utilization. Front. Pharmacol. 5:105. doi: 10.3389/fphar.2014.00105
This article was submitted to Experimental Pharmacology and Drug Discovery, a
section of the journal Frontiers in Pharmacology.
Copyright © 2014 Mandal, Roy, Ghosh, Hazra, Basak and Franco. This is an open-
access article distributed under the terms of the Creative Commons Attribution License
(CC BY). The use, distribution or reproduction in other forums is permitted, provided
the original author(s) or licensor are credited and that the original publication in this
journal is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Frontiers in Pharmacology |Experimental Pharmacology and Drug Discovery May 2014 |Volume 5 |Article 105 |12
... Combination therapy: Combining multiple antibiotics with different mechanisms of action can help overcome resistance mechanisms and improve treatment outcomes. Non-conventional alternative treatment strategies: Exploration of the possibility of using phage therapy ( Fig. 6), vaccination (as stated earlier), natural or synthetic antimicrobial peptides, and the use of AI to develop unique antibiotics are among the new regimes that could provide unique solutions in the future [167][168][169][170]. Nanorobotics technology is also a new area of research that could be investigated for its benefits in this challenge. ...
... Integrated Approach: A comprehensive approach that considers both the benefits and risks of B. cepacia is necessary for its responsible use and effective management. In essence, B. cepacia presents a complex challenge [169,170] with both positive and negative aspects. Recognizing its potential benefits while prioritizing the mitigation of health risks is crucial for navigating its role in our world. ...
Article
Full-text available
This review explores the paradox of Burkholderia species, a bacterium capable of both beneficial and harmful actions. Genus Burkholderia is a Gram-negative, rod-shaped, motile bacterium with remarkable genetic diversity. It can thrive in diverse environments and exchange genes with other bacteria through horizontal gene transfer. The ambivalence of Burkholderia lies in its potential to act as both a plant pathogen and an opportunistic human pathogen. Early isolations linked it to urinary tract infections and meningitis. However, its true threat emerged for individuals with cystic fibrosis and chronic granulomatous disease, who are susceptible to severe Burkholderia infections like pneumonia and septicemia. Beyond healthcare, Burkholderia exhibits useful applications in agriculture and biotechnology. Certain strains promote plant growth by fixing nitrogen, solubilizing phosphorus, and producing growth hormones. Burkholderia offers promise in bioremediation, degrading pollutants like hydrocarbons, pesticides and heavy metals. However, responsible application is crucial. Environmental contamination, biofilm formation, and potential plant diseases raise concerns. Careful consideration and ongoing research are necessary to mitigate these risks. Biotechnology leverages Burkholderia's diverse functionalities. It produces bioplastics, enzymes, and rhamnolipids with potential applications in various industries. Despite its benefits, it poses a healthcare risk to immunocompromised individuals, causing respiratory infections and hospital outbreaks. Its multi-drug resistance necessitates strict infection control protocols and development of novel treatment strategies. In conclusion, Burkholderia presents a complex picture. While its potential benefits in agriculture and biotechnology are promising, responsible use and continuous research are paramount to exploit its advantages and mitigate potential hazards.
... This characteristic enables AMPs to adhere to and infiltrate the bacterial cell wall bilayer, resulting in the formation of pores through mechanisms known as "toroidal-pore," "barrel-stave," and "carpet". Consequently, this process leads to the leakage of intracellular contents [81]. They come in a variety of structural forms including helical to linear and β-sheet structures (Fig. 4) [82]. ...
... The mechanisms for intracellular AMPs are summarized in Fig. 6. The antimicrobial activity of AMPs is particularly linked with its corresponding amino acid composition and physicochemical characteristics [81]. In addition to their direct antimicrobial activity, AMPs possess immunomodulatory properties, which stimulate the immune reaction of the host animal. ...
Article
Full-text available
Globally, Mastitis is a disease commonly affecting dairy cattle which leads to the use of antimicrobials. The majority of mastitis etiological agents are bacterial pathogens and Staphylococcus aureus is the predominant causative agent. Antimicrobial treatment is administered mainly via intramammary and intramuscular routes. Due to increasing antimicrobial resistance (AMR) often associated with antimicrobial misuse, the treatment of mastitis is becoming challenging with less alternative treatment options. Besides, biofilms formation and ability of mastitis-causing bacteria to enter and adhere within the cells of the mammary epithelium complicate the treatment of bovine mastitis. In this review article, we address the challenges in treating mastitis through conventional antibiotic treatment because of the rising AMR, biofilms formation, and the intracellular survival of bacteria. This review article describes different alternative treatments including phytochemical compounds, antimicrobial peptides (AMPs), phage therapy, and Graphene Nanomaterial-Based Therapy that can potentially be further developed to complement existing antimicrobial therapy and overcome the growing threat of AMR in etiologies of mastitis.
... Issue) : 2024 healthy medicinal plant extracts comprising high therapeutic values (Satyavati et al., 1987;Sandhya et al., 2006). The serious impact of anti-microbial resistance (AMR) pathogens has spurred researchers to discover extra productive novel drugs as an alternative source to safeguard human beings their (Mandal et al. 2014). Modification in the structural and functional mechanism of microbes strengthens its resistance potency which facilitated the evolution of antibiotic-resistant microbial strains such as Methicillin-Resistant Staphylococcus aureus (MRSA), Extended-Spectrum Beta-Lactamases (ESBL), Klebsiella ST258 and Escherichia coli ST131. ...
... Esto es posible dado que los fagos expresan genes que pueden inhibir el crecimiento de las bacterias, las cuales pueden ser patogénicas para sus respectivos hospederos, como en el caso del hombre. El uso de los de los fagos como alternativa al uso de antibióticos convencionales ha sido explorada desde hace años y es conocida como fagoterapia (Sulakvelidze et al., 2001;Hagens et al., 2004;Harper & Enright, 2011;Mandal et al., 2014;Kaur, 2016). En este sentido, el modelo de D. melanogaster también ha sido empleado para evaluar la eficacia antibacterial de los fagos contra un patógeno humano oportunista, como lo es Pseudomonas aeruginosa (Heo et al., 2009;Jang et al., 2018). ...
Article
El uso de los de los fagos como alternativa al uso de antibióticos convencionales ha sido explorada desde hace años y es conocida como fagoterapia (Sulakvelidze et al., 2001; Hagens et al., 2004; Harper & Enri¬ght, 2011; Mandal et al., 2014; Kaur, 2016). En este sentido, el modelo de D. melanogaster también ha sido empleado para evaluar la eficacia antibacterial de los fagos contra un patógeno humano oportunista, como lo es Pseudomonas aeruginosa (Heo et al., 2009; Jang et al., 2018). Esto es sólo una muestra de un enfoque que promete arrojar más información sobre el uso potencial de los fagos, aunque, y todavía más importante, también representa un nuevo punto de partida para identificar e inda¬gar sobre interacciones todavía no descritas. Si un solo modelo como D. melanogaster ha sido capaz de aportar tanto a la biología, ahora falta esperar qué nos puede decir ésta en conjunto a sus bacterias y virus en conjunto.
... At 5 min, the disappearance of peaks at 657 and 1096 cm −1 , assigned to DNA, indicates that the host DNA is degraded. 68 Moreover, from 5 min onward, the biosynthesis of phage DNA and proteins takes place, 68 which is characterized by the appearance of peaks at 620, 743, 1113 and 1273 cm −1 (proteins) and the shi of the SERS peak assigned to DNA at 728 cm −1 (control) to 732 cm −1 (different time intervals). In addition to this, the appearance of the SERS peaks at 1331 cm −1 (assigned to DNA) and 1454 cm −1 (lipids) indicates the bacteriophage replication in the host bacteria. ...
Article
Full-text available
Drug-resistant pathogenic bacteria are a major cause of infectious diseases in the world and they have become a major threat through the reduced efficacy of developed antibiotics. This issue can be addressed by using bacteriophages, which can kill lethal bacteria and prevent them from causing infections. Surface-enhanced Raman spectroscopy (SERS) is a promising technique for studying the degradation of infectious bacteria by the interaction of bacteriophages to break the vicious cycle of drug-resistant bacteria and help to develop chemotherapy-independent remedial strategies. The phage (viruses)-sensitive Staphylococcus aureus (S. aureus) bacteria are exposed to bacteriophages (Siphoviridae family) in the time frame from 0 min (control) to 50 minutes with intervals of 5 minutes and characterized by SERS using silver nanoparticles as SERS substrate. This allows us to explore the effects of the bacteriophages against lethal bacteria (S. aureus) at different time intervals. The differentiating SERS bands are observed at 575 (C–C skeletal mode), 620 (phenylalanine), 649 (tyrosine, guanine (ring breathing)), 657 (guanine (COO deformation)), 728–735 (adenine, glycosidic ring mode), 796 (tyrosine (C–N stretching)), 957 (C–N stretching (amide lipopolysaccharides)), 1096 (PO2 (nucleic acid)), 1113 (phenylalanine), 1249 (CH2 of amide III, N–H bending and C–O stretching (amide III)), 1273 (CH2, N–H, C–N, amide III), 1331 (C–N stretching mode of adenine), 1373 (in nucleic acids (ring breathing modes of the DNA/RNA bases)) and 1454 cm⁻¹ (CH2 deformation of saturated lipids), indicating the degradation of bacteria and replication of bacteriophages. Multivariate data analysis was performed by employing principal component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA) to study the biochemical differences in the S. aureus bacteria infected by the bacteriophage. The SERS spectral data sets were successfully differentiated by PLS-DA with 94.47% sensitivity, 98.61% specificity, 94.44% precision, 98.88% accuracy and 81.06% area under the curve (AUC), which shows that at 50 min interval S. aureus bacteria is degraded by the replicating bacteriophages.
... Antimicrobial peptides (AMPs) have been considered as an alternative therapeutic agent to conventional antibiotics [10,11]. These naturally occurring peptides, typically consisting of less than 100 hydrophobic and cationic amino acids in a certain sequence combination, exert their antibacterial effects mainly by disrupting the integrity of bacterial cell membranes [12]. ...
Article
Full-text available
Rationale: Antimicrobial peptide LL-37 has been recognized as a favorable alternative to antibiotics due to its broad antibacterial spectrum, low resistance development and diverse biological activities. However, its high manufactory cost, poor proteolytic stability, and unpredictable cytotoxicity seriously hindered its medical translation. Methods: To push the frontiers of its clinical application, all-hydrocarbon stapling strategy was exploited here for the structural modification of KR-12, the core and minimal fragment of LL-37. Results: Based on a library of KR-12 derivatives that designed and synthesized to be stapled at positions of either i, i+4 or i, i+7, structure to activity relationship was investigated. Among them, KR-12(Q5, D9) with the glutamine and aspartic acid residues stapled displayed increased helical content and positive charge. The reinforced α-helical conformation not only protected it from proteolytic hydrolysis but also improved its antibacterial efficacy via effective membrane perturbation and anti-inflammatory efficacy via compact LPS binding. Besides, the increased positive charge endowed it with an enhanced therapeutic index. On infected wound mouse model, it was demonstrated to eliminate bacteria and promote wound closure and regeneration effectively. Conclusion: Overall, the all-hydrocarbon stapling was proven to lay the foundation for the future development of antibacterial agents. KR-12(Q5, D9) could serve as a lead compound for the clinical treatment of bacterial infections.
Chapter
Nowadays, one of the major challenges in biomedical and biopharmaceutical field is designing novel and effective anti-amyloidogenic inhibitors for the treatment of various human pathophysiologies associated with protein aggregation. In this milieu, numerous small molecules, polyphenols, surfactants, nanoparticles, etc. have been extensively studied to explore their anti-amyloidogenic properties, and thus provide huge scope for them to appear as future therapeutic agents in the treatment of amyloidogenic disorders. Recently, inspired by the fascinating properties of polymers such as non-toxicity, excellent biocompatibility, tuneable architectures, controllable degradation rate, possibility of multiple interaction between amyloidogenic protein/peptide and polymer, and excellent in vivo stability, polymer-based therapeutic agents have been extensively explored in the field of protein misfolding and aggregation. This mini-review article emphasizes the recent advancements of polymeric materials in the field of protein aggregation for ameliorating neurodegenerative diseases. Finally, we conclude this mini-review by providing some viewpoints on future directions
Article
This research presents an innovative method for biosynthesizing vanadium oxide rod-like nanostructures leveraging a Ganoderma lucidum (mushroom) plant extract. By harnessing specific extracts from Ganoderma lucidum, the procedure uniquely employed them as both reducing and capping agents during the nanoparticle formation. These synthesized nanoparticles were then subjected to a detailed characterization using various techniques, such as X-ray Diffraction (XRD), Fourier Transform Infrared Spectroscopy (FTIR), X-ray Fluorescence (XRF), Dynamic Light Scattering (DLS), and Scanning Electron Microscopy (SEM). The FTIR analysis verified the synthesis of the vanadium oxide nanoparticles, with surface capping likely due to phytochemical groups from the plant extract. Additional authentication came from the XRF analysis, which confirmed the significant presence of vanadium, further emphasizing their composition. DLS results showed a favorable average particle size of 57.55 nm and a polydispersity index of 0.356, hinting at a well-distributed size range. Through XRD analysis, a well-defined crystal structure and atomic arrangement were identified for the vanadium nanoparticles, evident from the specific diffraction peaks. Importantly, the SEM study shed light on their unique rod-like morphology—elongated structures of consistent diameter and defined length—underscoring their impressive high aspect ratio. Antimicrobial assessments revealed variable effects against different bacterial strains. While vanadium oxide nanoparticles demonstrated promising inhibitory action against Klebsiella pneumoniae at specific concentrations, they exhibited limited activity against Staphylococcus aureus and Escherichia coli across the studied concentration range. Considering their distinct morphology and the potential antimicrobial properties, these nanoparticles present a rich prospect for a multitude of applications. Their potential extends beyond just antimicrobial uses and may span areas such as catalysis, energy storage, sensing, and other advanced technological applications.
Article
In the food industry, despite the widespread use of interventions such as preservatives and thermal and non-thermal processing technologies to improve food safety, incidences of foodborne disease continue to happen worldwide, prompting the search for alternative strategies. Bacteriophages, commonly known as phages, have emerged as a promising alternative for controlling pathogenic bacteria in food. This review emphasizes the potential applications of phages in biological sciences, food processing, and preservation, with a particular focus on their role as biocontrol agents for improving food quality and preservation. By shedding light on recent developments and future possibilities, this review highlights the significance of phages in the food industry. Additionally, it addresses crucial aspects such as regulatory status and safety concerns surrounding the use of bacteriophages. The inclusion of up-to-date literature further underscores the relevance of phage-based strategies in reducing foodborne pathogenic bacteria's presence in both food and the production environment. As we look ahead, new phage products are likely to be targeted against emerging foodborne pathogens. This will further advance the efficacy of approaches that are based on phages in maintaining the safety and security of food.
Article
Full-text available
Comparative genomics demonstrated that the chromosomes from bacteria and their viruses (bacteriophages) are coevolving. This process is most evident for bacterial pathogens where the majority contain prophages or phage remnants integrated into the bacterial DNA. Many prophages from bacterial pathogens encode virulence factors. Two situations can be distinguished: Vibrio cholerae, Shiga toxin-producing Escherichia coli, Corynebacterium diphtheriae, and Clostridium botulinum depend on a specific prophage-encoded toxin for causing a specific disease, whereas Staphylococcus aureus, Streptococcus pyogenes, and Salmonella enterica serovar Typhimurium harbor a multitude of prophages and each phage-encoded virulence or fitness factor makes an incremental contribution to the fitness of the lysogen. These prophages behave like "swarms" of related prophages. Prophage diversification seems to be fueled by the frequent transfer of phage material by recombination with superinfecting phages, resident prophages, or occasional acquisition of other mobile DNA elements or bacterial chromosomal genes. Prophages also contribute to the diversification of the bacterial genome architecture. In many cases, they actually represent a large fraction of the strain-specific DNA sequences. In addition, they can serve as anchoring points for genome inversions. The current review presents the available genomics and biological data on prophages from bacterial pathogens in an evolutionary framework.
Article
Gene-encoded peptide antibiotics are ubiquitous components of host defenses in mammals, birds, amphibia, insects, and plants. Their de novo synthesis or release from storage sites can be induced rapidly, which makes them particularly important in the initial phases of resistance to microbial invasion. The endogenous antimicrobial peptides of animals are products of single genes and are synthesized as preproproteins. Multistep processing yields the mature peptide, which generally acts by inducing microbial membrane permeabilization. Several families of antimicrobial peptides have been identified that differ with respect to the presence of disulfide linkages, amino acid composition, structural conformation, and spectrum of activity. The arginine-rich three disulfide-containing β-βheet defensins are remarkably abundant and widely distributed in animals and plants. The antibiotic peptides of higher eukaryotes merit further study for their role in natural immunity and their potential as novel therapeutic compounds. J. Leukoc. Biol. 58: 128–136; 1995.
Article
The increasing resistance of bacteria to conventional antibiotics resulted in a strong effort to develop antimicrobial compounds with new mechanisms of action. Antimicrobial peptides seem to be a promising solution to this problem. Many studies aimed at understanding their mode of action were described in the past few years. The most studied group includes the linear, mostly α-helical peptides. Although the exact mechanism by which they kill bacteria is not clearly understood, it has been shown that peptide–lipid interactions leading to membrane permeation play a role in their activity. Membrane permeation by amphipathic α-helical peptides can proceed via either one of the two mechanisms: (a) transmembrane pore formation via a “barrel-stave” mechanism; and (b) membrane destruction/solubilization via a “carpet-like” mechanism. The purpose of this review is to summarize recent studies aimed at understanding the mode of action of linear α-helical antimicrobial peptides. This review, which is focused on magainins, cecropins, and dermaseptins as representatives of the amphipathic α-helical antimicrobial peptides, supports the carpet-like rather the barrel-stave mechanism. That these peptides vary with regard to their length, amino acid composition, and net positive charge, but act via a common mechanism, may imply that other linear antimicrobial peptides that share the same properties also share the same mechanism. © 1999 John Wiley & Sons, Inc. Biopoly 47: 451–463, 1998
Article
Prodrugs are bioreversible derivatives of drug molecules used to overcome some barriers to the utility of the parent drug molecule. These barriers include, but are not limited to, solubility, permeability, stability, presystemic metabolism and targeting limitations. The patent literature shows a dramatic increase in numbers of prodrug patents (> 2000% increase in 2002 compared to 1993), with claims for cancer treatment comprising 37% of these. This increase is largely due to the rise from North American-based multinationals and some smaller drug delivery companies mirroring the overall trend. In 2001 and 2002, 14% of all new approved chemical drugs were prodrugs. It appears that prodrugs to overcome barriers to the delivery of problematic drug candidates are becoming an integral part of the drug discovery paradigm.
Article
Antibiotic peptides are a key component of the innate immune systems of most multi-cellular organisms. Despite broad divergences in sequence and taxonomy most antibiotic peptides shale a common mechanism of action, i.e., membrane permeabilization of the pathogen. This review provides a general introduction to the subject, with emphasis on aspects such as structural types, post-translational modifications, mode of action or mechanisms of resistance. Some of these questions are treated in depth in other reviews in this issue. The review also discusses the role of antimicrobial peptides in nature, including several pathological conditions, as well as recent accounts of their application at the preclinical level. (C) 1999 John Wiley & Sons, Inc.
Article
We have previously described a family of cationic amphipathic peptides derived from lentivirus envelope proteins that have properties similar to those of naturally occurring antimicrobial peptides. Here, we explored the effects of amino acid truncations and substitutions on the antimicrobial potency and selectivity of the prototype peptide, LLP1. Removal of seven residues from the C-terminus of LLP1 had Ilttle effect on potency, but abrogated haemolytic activity. Replacement of the two glutamic acid residues of LLP1 with arginine resulted in a peptide with greater bactericidal activity. We discovered that the cysteine-containing peptides spontaneously formed disulphide-linked dimers, which were 16-fold more bactericidal to Staphylococcus aureus. Monomeric and dimeric LLP1 possessed similar alpha helical contents, indicating that disulphide formation did not alter the peptide's secondary structure. The dimerization strategy was applied to magainin 2, enhancing its bactericidal activity eight-fold. By optimizing all three properties of LLP1, a highly potent and selective peptide, named TL-1, was produced. This peptide Is significantly more potent than LLP1 against Gram-positive bacteria while maintaining high activity against Gram-negative organisms and low activity against eukaryotic cells. In addition to new antimicrobial peptides, these studies contribute useful information on which further peptide engineering efforts can be based.