712
Views
291
CrossRef citations to date
0
Altmetric
Review

Nanomaterials for alternative antibacterial therapy

Pages 8211-8225 | Published online: 10 Nov 2017

Abstract

Despite an array of cogent antibiotics, bacterial infections, notably those produced by nosocomial pathogens, still remain a leading factor of morbidity and mortality around the globe. They target the severely ill, hospitalized and immunocompromised patients with incapacitated immune system, who are prone to infections. The choice of antimicrobial therapy is largely empirical and not devoid of toxicity, hypersensitivity, teratogenicity and/or mutagenicity. The emergence of multidrug-resistant bacteria further intensifies the clinical predicament as it directly impacts public health due to diminished potency of current antibiotics. In addition, there is an escalating concern with respect to biofilm-associated infections that are refractory to the presently available antimicrobial armory, leaving almost no therapeutic option. Hence, there is a dire need to develop alternate antibacterial agents. The past decade has witnessed a substantial upsurge in the global use of nanomedicines as innovative tools for combating the high rates of antimicrobial resistance. Antibacterial activity of metal and metal oxide nanoparticles (NPs) has been extensively reported. The microbes are eliminated either by microbicidal effects of the NPs, such as release of free metal ions culminating in cell membrane damage, DNA interactions or free radical generation, or by microbiostatic effects coupled with killing potentiated by the host’s immune system. This review encompasses the magnitude of multidrug resistance in nosocomial infections, bacterial evasion of the host immune system, mechanisms used by bacteria to develop drug resistance and the use of nanomaterials based on metals to overcome these challenges. The diverse annihilative effects of conventional and biogenic metal NPs for antibacterial activity are also discussed. The use of polymer-based nanomaterials and nanocomposites, alone or functionalized with ligands, antibodies or antibiotics, as alternative antimicrobial agents for treating severe bacterial infections is also discussed. Combinatorial therapy with metallic NPs, as adjunct to the existing antibiotics, may aid to restrain the mounting menace of bacterial resistance and nosocomial threat.

Introduction

Hospital-acquired bacterial infections, mainly caused by the nosocomial pathogens such as Acinetobacter baumannii, Pseudomonas aeruginosa, Escherichia coli, Staphylococcus aureus, Klebsiella pneumoniae and so on, pose the foremost challenge to the well-being of a patient.Citation1 The bacteria counteracts the host’s innate immune defense machinery,Citation2,Citation3 which becomes the prime cause of death in patients confined to the intensive care unit (ICU), with weakened immune system, culminating in invasive bloodstream infections. The widespread use of broad-spectrum antibioticsCitation4 has led to the appearance of multidrug-resistant (MDR) isolates that further intricate the clinical problem as the bacteria spread epidemically among the patients. With the compromising efficacy of the available chemotherapeutics due to mounting drug resistance and the biofilm recalcitrance towards antibiotics, there is a pressing need to identify alternate drugs. In this respect, nanomaterials have shown promise owing to their unique physical and chemical attributes.Citation5Citation7 Their large surface area relative to volume enables intimate interactions with microbial membranes, as well as surface functionalization, which help in developing more effective antibacterial agents. Over the last decade, there has been a remarkable global focus on conventional as well as biogenic metallic nanoparticles (NPs) as innovative tools for combating the high rates of antimicrobial resistance. Chemotherapeutic drugs when given in combination with metallic NPs may result in a cumulative effect due to the antibiotic as well as the metal ions released from NPs. Moreover, the antibacterial agent may be used at a much lower dose than when administered alone, hence overcoming the problem of resistance and diminishing other undesirable side effects to some extent.Citation6,Citation8 There has also been a paradigm shift in management of biofilms and MDR bacteria with polymeric nanocomposites and antibiotic-loaded polymeric NPs. Improved therapeutic efficacy with concomitant decline in side effects of antimicrobial drugs has also been achieved by surface modification of metallic NPs with ligands or antibodies for targeted delivery.

This review summarizes the immune evasion strategies and antibiotic resistance mechanisms employed by bacteria to survive in the host and the probable metallic nanomaterials-based bactericidal effects to fight against nosocomial pathogens. The antibacterial activities of biologically synthesized metallic NPs as well as polymeric nanocomposites and surface-modified NPs are also highlighted. The metal-based nanomaterials alone or functionalized with antibiotics when translated to clinics may show promise as next-generation nanotherapeutics against bacterial menace.

Gravity of the problem

Bacterial infections have emerged as the leading cause of the formidable rates of deaths in hospitalized and immunosuppressed patients, especially those in ICUs and those undergoing invasive operations, worldwideCitation1,Citation4,Citation9Citation11 as well as in parts of Saudi Arabia where the prevalence has been reported to be 31.7%.Citation12 The hospital-acquired infections manifest in a wide gamut of severe clinical ramifications, such as bacteremia, septic shock, ventilator-related pneumonia and massive soft tissue necrosis, and rapidly progress to systemic infections that eventually culminate in multiorgan failure and death.Citation13 The control measures including implementation of hygiene, patient isolation and environmental decontamination have proved ineffective in keeping the infection at bay. Improper use of antibiotics has favored an upward trend in the development of resistance to almost all the available drugs, further compounding the clinical problem.Citation9,Citation14 The challenge to control these infections is augmented in MDR bacteria such as those producing extended-spectrum β-lactamases and carbapenemases (K. pneumoniae carbapenase) and in methicillin-resistant S. aureus (MRSA). Drug combination regimens have also proved ineffective due to formation of biofilms, agglomerates of bacterial colonies that adhere to a surface and resist traditional means of killing by avoiding contact with the antibiotics.Citation15 The bacteria survive in the biofilms for extended periods of time and are likely to be transmitted within the health care settings. The quandary of mechanisms of antibiotic resistance has retrogressed the clinical outcome and inflated the economic burden of infectious diseases, leaving the medical practitioners with few therapeutic options to address the emerging threat.Citation14 Furthermore, an arsenal of strategies has been employed by bacteria to subvert the host immune system, adversely impacting the surrogate markers of clinical course such as the length of hospitalization and hospital-related deaths.

Bacteria’s stratagem: targeting the innate immune defense

The bacteria have evolved a diverse array of resistance mechanisms to disable key players of the host innate immune defenses for their survival (). They utilize a multitude of virulence factors that conjointly render substantial impediment to phagocytes (macrophages and neutrophils) recruitment and activation.Citation3 The bacteria parasitize host cells by arresting or reprogramming phagosomal maturation, by escaping maturing phagosomes or by withstanding the microbicidal properties of the phagolysosome. They acquire resistant proteins to withstand low pH environment of phagosomes. The bacteria escape neutrophils extracellular traps (NETs) and elude opsonophagocytosis, and the cytocidal effects of host’s antimicrobial peptides (AMPs) and reactive oxygen species (ROS). They secrete proteases that cleave NETs, degrade AMPs and express detoxifying enzymes like catalase to neutralize ROS and convert them into less harmful compounds.Citation16 Bacteria may also impair ROS production. Bacteria trigger phagocytic death through pore-forming cytolysins and escape host complement deposition and activation.Citation2 They release complement-degrading and complement-inactivating enzymes and inhibit membrane attack complex polymerization, thereby impeding bacteriolysis. Self-colonization of bacteria to form biofilms further leads to immune subterfuge.Citation6 Bacteria subvert the T cell stimulation by dendritic cells (DCs), through downregulation of their antigen-presenting or costimulatory functions. Bacteria may directly infect DC, dampening its interleukin (IL)-12 and tumor necrosis factor-alpha (TNF-α) production, thereby crippling the adaptive immune response.Citation17 Immune evasion favors bacterial persistence, resulting in increased antibiotic use and selection for MDR pathogens, thus emphasizing the need for alternate antimicrobial therapies.Citation16

Figure 1 Strategies for survival in the host to spark invasive infections. (A) Innate immune mechanisms evaded by bacteria include phagocyte (macrophages, neutrophils) recruitment and activation, opsonization via Fc receptors on macrophages, complement activation and the bactericidal activities of antimicrobial peptides and reactive oxygen species. (B) Drug resistance mechanisms evolved by bacteria comprise hydrolysis by β-lactamases, modification of drug targets or antibiotics, loss or mutation of porins and overexpression of efflux pumps.

Abbreviations: AMP, antimicrobial peptide; ROS, reactive oxygen species; AME, aminoglycoside-modifying enzyme; MDR, multidrug resistant; LPS, lipopolysaccharide.

Figure 1 Strategies for survival in the host to spark invasive infections. (A) Innate immune mechanisms evaded by bacteria include phagocyte (macrophages, neutrophils) recruitment and activation, opsonization via Fc receptors on macrophages, complement activation and the bactericidal activities of antimicrobial peptides and reactive oxygen species. (B) Drug resistance mechanisms evolved by bacteria comprise hydrolysis by β-lactamases, modification of drug targets or antibiotics, loss or mutation of porins and overexpression of efflux pumps.Abbreviations: AMP, antimicrobial peptide; ROS, reactive oxygen species; AME, aminoglycoside-modifying enzyme; MDR, multidrug resistant; LPS, lipopolysaccharide.

The emergence of antibiotic resistance

In the present era, the quintessential antibiotics remain the mainstay for management of invasive infections, to annihilate or restrain the growth of a vast spectrum of bacteria. However, this perspective advocates ever-expanding threats accountable to indiscriminate use of antimicrobials resulting in escalated incidence of antibiotic resistance and epidemics of hypervirulent pathogens among vulnerable patients.Citation14 Bacteria have progressively eroded the effectiveness of not only a single antibiotic by developing resistance but concurrently multiple drugs that were previously effective. Intracellular bacteria are difficult to treat with regular antimicrobial therapies, resulting in chronic infections. Medical devices such as catheters, implants and sutures increase the risks of nosocomial infections due to formation of biofilms. The biofilm-related infections are extremely defiant to antimicrobial arsenal and spread rampantly in the community.Citation15 The normal gut microbiota that otherwise maintains the immune functions is also targeted by antibiotic spectrum. Drug-resistant bacterial infections entail higher doses of drugs, with augmented toxicity, longer hospital stays and enhanced mortality. Thus, antimicrobial resistance remains a substantial global health concern, invigorating the critical need for alternate therapeutic options to combat chronic intracellular infections and biofilms so as to shorten the hospital stays, and hence mortality.

Drug resistance subterfuge evolved by bacteria

The acquisition of resistance to single and multiple antibiotics by bacteria () has been reported to occur through horizontal gene transfer on plasmids or transposons by transformation, conjugation and transduction or by spontaneously mutating the existing genes.Citation18 The resistance becomes rampant due to selective pressure for microbes expressing resistance genes against the antimicrobial drug, created by overuse of antibiotics, non-patient compliance or use of time-dependent drugs with long half-lives or microbiostatic drugs.Citation7 Modes of resistance mechanismsCitation19 that endanger the efficacy of antibiotic arsenal are summed up as follows.

Active antimicrobial drug efflux and reduced uptake

Drug efflux pumps preclude the entry of drugs or extrude the antibacterial agent from the microbial cell before it reaches its target site to exert its effect. Overexpression of transmembrane multidrug efflux pumps as well as reduced uptake results in sub-toxic levels of drugs within the microbial cells.Citation20 In P. aeruginosa, mutation in regulatory protein that normally dampens the gene encoding efflux proteins results in enhanced outpour. E. coli uses transmembrane proton gradient to expel multiple antibiotics through its numerous efflux pumps. The increased drug resistance of Gram-positive bacteria is also attributed to the outer membrane surrounding the periplasmic space that restrains the uptake of hydrophobic drugs. The antibiotic uptake or efflux may also be affected by mutations such as the diminished expression or absence of porins in P. aeruginosa, lowering the permeability of the cell wall to carbapenems.Citation21 In case of both Gram-positive and Gram-negative bacteria, one of the genes encoding tetracycline efflux pumps (TetA) is normally not expressed due to TetR repressor protein. However, tetracycline binds to and inactivates TetR, thereby inducing TetA expression that catalyzes drug efflux.Citation7

Expression of resistance gene encoding altered substrate

The antibacterial agent has diminished affinity for the resistance gene-encoded altered antibiotic target sites on the substrate than the wild-type site. This is exemplified in case of penicillin-resistant Streptococcus pneumoniae (due to MecA gene conferring resistance to all β-lactams), vancomycin-resistant S. aureus and vancomycin-resistant Enterococcus (due to VanA resistance gene), sulfonamide-resistant S. pneumoniae, Streptococcus pyogenes, Neisseria spp. and E. coli.Citation7 This mechanism also accounts for conferring resistance against aminoglycosides, macrolides, β-lactams, sulfonamides, linezolid, tetracyclines, rifampin and quinolones.Citation7,Citation22 Resistance genes coding for enzymes that methylate 23S rRNA of the 50S ribosomal subunit and 16S rRNA of the 30S ribosomal subunit inhibit binding by macrolides (such as erythromycin) and aminoglycosides, respectively.Citation7,Citation23

Covalent modification of antimicrobial drug, dampening its efficacy

Plasmids or transposons and rarely bacterial chromosomes harbor resistance genes encoding aminoglycoside-modifying enzymes that covalently alter the OH or NH2 groups on aminoglycosides, thereby undermining their affinity for 30S ribosomal subunit, and waning their antimicrobial activity. β-Lactamases, having broad-spectrum activity against most of the β-lactam antibiotics, including cephalosporins, have evolved by gene transfer.Citation5,Citation21 Chloramphenicol acetyl-transferases modify chloramphenicol, making it unable to bind to 50S ribosomal subunit and inhibit protein synthesis. Covalent modification also affords resistance against tobramycin, gentamycin, chloramphenicol, kanamycin, macrolides, tetracyclines, quinolones and streptogramins.Citation7,Citation23

Synthesis of a competitive inhibitor of antibiotic

S. aureus and Neisseria meningitidis acquire sulfonamide resistance by producing its inhibitor (para-aminobenzoic acid) that competes for binding the active site of bacterial dihydropteroate synthetase.Citation7,Citation24 Mutations in this enzyme have also been found in many clinical isolates, downgrading sulfonamide-based therapies to second- or third-line options.

Biofilm formation to avoid contact with antibiotics

Biofilms are surface-adherent aggregates of bacterial communities embedded within an extracellular, self-produced, polymeric matrix. Intracellular bacteria in biofilms have limited exposure to antibiotics. Biofilms also act as a barrier of diffusion by trapping and degrading antibiotics and thus render tolerance to even high concentrations of antibiotics, a phenomenon called recalcitrance, resulting in recurrent or chronic bacterial infections as with S. aureus and P. aeruginosa.Citation15 Biofilms can also favor gene transfer between bacteria, thus spreading antibiotic resistance and transforming a previously non-virulent commensal into a virulent pathogen.Citation25

Nanotechnology-based therapeutic interventions to fight nosocomial pathogens

Several metals, metal oxides, metal halides and bimetals in nanoparticulate form have been documented for antimicrobial activityCitation5,Citation26 as the bacteria are less likely to develop resistance to nanomaterials. These comprise NPs containing Ag, Au, Zn, Cu, Ti, Mg, Ni, Ce, Se, Al, Cd, Y, Pd and super-paramagnetic Fe. Zerovalent bismuth-containing NPs have shown promise in treating infections due to drug-resistant bacteria in combination with X-rays.Citation27 Among the metal-containing NPs, Au NPs have moderate antibacterial activity unless their surface is modified. Ag NPs are the most effective nano-weapon against bacterial infections.Citation28Citation30 On the other hand, upsurge in resistance to Ag NPs has been reported due to genetic alterations in bacteria.Citation31 The deposition of silver of Ag NPs in the liver, spleen, lungs and other organs results in organ damage and dysfunction, and seriously erodes its efficacy. Al2O3 NPs promote horizontal conjugative transfer of MDR genes, increasing antibiotic resistance.Citation32 The high toxicity of CuO NPs causes oxidative lesions, while DNA damage induced by ZnO and TiO2 NPs limits the efficacy of these NPs. Nonetheless, NPs have emerged as alternative antimicrobial approach to combat biofilms and for treating severe bacterial infections.Citation33 The NPs-mediated elimination of the microbes may be microbicidal, or the effect may be microbiostatic, wherein the growth of bacteria is arrested and metabolic activities are ceased and the killing is potentiated by the host’s immune cells.

Mode of action of metal-based nanomaterials

Metallic NPs use multifaceted contrivance simultaneously to combat microbes (), depreciating the probability of development of resistance, as it would entail multiple concurrent gene mutations in the same microbe for evolvement of that resistance. The molecular mechanisms by which metal-based NPs annihilate MDR bacteria, resulting in disturbance in respiration and inhibition of cellular growth, have been extensively reviewed.Citation28,Citation29,Citation33 summarizes the metallic NPs that have been reported for antibacterial activity via an array of mechanisms enlisted as follows.

Table 1 Metallic nanomaterials-based probable bactericidal effects

Figure 2 Probable nanomaterials-based bactericidal effects. Nanomaterials trigger release of heavy metal ions that intercalate between bases, damage cellular proteins, disrupt cell signaling, generate free radicals and prevent biofilm formation.

Abbreviation: ROS, reactive oxygen species.

Figure 2 Probable nanomaterials-based bactericidal effects. Nanomaterials trigger release of heavy metal ions that intercalate between bases, damage cellular proteins, disrupt cell signaling, generate free radicals and prevent biofilm formation.Abbreviation: ROS, reactive oxygen species.

ROS generation

The toxicity of nanomaterials has been attributed to ROS production such as hydroxyl radicals, superoxide anions and hydrogen peroxide that inhibit DNA replication as well as amino acid synthesis and damage the bacterial cell membranes via lipid peroxidation, compromising membrane semipermeability and repressing oxidative phosphorylation. Hydroxyl radical (•OH) formation has been observed with Ag NPsCitation28,Citation29,Citation34 and hydrogen peroxide (H2O2) with ZnO NPs,Citation35 while TiO2 NPsCitation36 produce both via photocatalysis. Free Cu++ from Cu-containing NPsCitation37 and Mg halogen (MgX2)-containing NPsCitation38 also induce formation of ROS.

Release of metal ions and disorganization of bacterial membrane

Different microorganisms have varying sensitivities to metal ions. Ag and ZnO NPs have been reported to exert antibacterial activity by release of Ag+ and Zn++ that disrupt the membrane.Citation28,Citation29,Citation35,Citation39 The antibacterial action of Ag NPs is revealed by interaction of Ag+ with sulfhydryl groups in enzymes and other cellular constituents, making them dysfunctional. Ag+ also precludes cell wall synthesis in Gram-positive bacteria. Cu++ interacts with amine and carboxyl groups on the surfaces of microbial cells, such as Bacillus subtilis.Citation40 Au NPs also result in bacterial membrane disruption.

Intercalation between DNA bases

Ag+ released from NPs has also been reported to interact with DNA of microbes,Citation28,Citation29 and inhibit DNA replication and cell division. Inhibition of DNA replication and DNA degradation has also been reported with Cu NPs.Citation37,Citation40 Bi NPs in combination with X-ray treatment emit electrons, with formation of free radicals that damage bacterial DNA.Citation48

Adsorption of nanomaterials to bacterial cell

The electrostatic interaction between the NPs and the microbial cells affects its toxicity, with the positively charged NPs being more toxic as has been observed with TiO2 and surface-modified Au NPs.Citation36,Citation41 The NPs adsorption to the surface of bacteria results in oxidative stress due to redox reactions, leading to toxicity. The adhesion of Ag and ZnO NPs to bacterial cell surface also results in damage to membrane lipids and proteins.Citation28,Citation29,Citation35

Alteration in bacterial membrane permeability

Cell surface adherence of Ag and polyvinyl alcohol (PVA)-coated ZnO NPs coupled with ROS generation increases membrane permeability and triggers cell death.Citation28,Citation29,Citation35 The membrane viscosity is also altered, influencing the transport across the membrane. Ag+ released from Ag NPs interacts with negatively charged lipopolysaccharide in the bacterial membrane, permeabilizing it and dissipating the electrochemical proton gradient across the membrane, resulting in lysis.Citation7 Ag+ inhibits cytochromes of the electron transport chain, and disrupt cellular transport systems by causing homeostatic imbalance due to K+ loss from the membrane. Loss of membrane potential and disruption of respiratory chain has also been observed with Au,Citation43 CuCitation37 and NiO NPs.Citation44

Penetration of the cell envelope and ribosome destabilization

The NP surface charge also affects the internalization and subcellular localization and hence toxicity as with CeCl2 NPs.Citation45 Entering of NPs into the cytoplasm to exert microbicidal effects via oxidative stress has been observed with Ag nanomaterialsCitation28,Citation29 and PVA-coated ZnO NPs.Citation35 MgF2 NPs cause lipid peroxidation and enter the membrane of microbial cells, causing a fall in cytoplasmic pH, which raises the membrane potential.Citation38 Al2O3 NPs cause oxidative damage to membrane and enter cytoplasm.Citation32 Ag and Au NPs have been reported to exert toxicity by penetrating inside the cell and denaturing 30S ribosomal subunit, thereby impeding protein translation.Citation29,Citation30,Citation41

Disruption of bacterial biofilms

During bacterial biofilm maturation, the extracellular matrix (slime) and extracellular carbohydrates known as quorum sensing molecules (for cell-to-cell communication) are produced.Citation46 The slow-growing bacterial cells detach, resulting in spread of infection. AgCitation28,Citation29 and ZnO NPsCitation47 are also documented to inhibit the microbial biofilm formation. YF2Citation48 and Se NPsCitation49 restrain growth and biofilm formation of E. coli and S. aureus. TiO2,Citation36 CdSCitation50, MgF2Citation38 and Bi NPsCitation27 have also been reported to disrupt bacterial biofilms. Magnetic NPs such as superparamagnetic iron oxide NPs coated with Ag or Au exhibit the greatest activity against bacterial biofilms.Citation39,Citation51

Antibacterial activities of ecofriendly green NPs

The potentiality of prokaryotes and plants to reduce inorganic metals has advanced a stimulating, cost-effective strategy towards the NPs synthesis via green nanotechnology.Citation57 Plant- as well as microbe-mediated metallic NPs synthesis (bottom-up or biological method) avoids the generation of toxic by-products, and is hence ecofriendly and a green alternative to conventional methods (top-down or physical methods and bottom-up or chemical methods) for mining nanomaterials.Citation58 The biologically synthesized metallic NPs are biocompatible and potentially safe for human therapeutic use. A plethora of literature highlights the antimicrobial activities of biogenic metallic NPs (). Rosmarinus officinalis leaf extract-mediated green synthesis of Ag NPs has been reported to have remarkable activity against B. subtilis, E. coli, P. aeruginosa and S. aureus,Citation59 while biogenic Ag NPs from Ficus benghalensis and Acalypha indica demonstrated antibacterial potential against B. subtilis, E. coli, P. aeruginosa and Vibrio cholerae.Citation60 Ag NPs synthesized using leaves extract of Skimmia laureola,Citation61 root extract of Delphinium denudatum,Citation62 Styrax benzoin extractCitation63 and Rosa chinensis flower extractCitation64 showed potent antibacterial effects against a spectrum of Gram-positive and Gram-negative human pathogenic bacteria. Ag NPs synthesized using Caesalpinia sappan extract served as a potential novel nanoantibiotic against MRSA.Citation65 Biologically synthesized Ag NPs using Coffea arabica seed extract have been found to exhibit significant antibacterial activity against E. coli and S. aureus, that was almost equivalent to that elicited with the standard drug ampicillin.Citation66 Inhibitory activity of green synthesized ZnO NPs from Solanum nigrum,Citation67 CeO2 NPs from Olea europaea leaf extract,Citation68 and Fe3O4-Ag core shell magnetic NPs obtained using Vitis vinifera stem extractCitation69 against both Gram-positive and Gram-negative pathogens has also been reported. The antibacterial activities of biologically synthesized NPs are due to the metal ions released from the NPs, coupled with the bio-organic compounds used in the green synthesis that may interact with the microbial membrane as well as preclude the need for reducing and stabilizing agent.Citation70 However, the biosynthetic NPs have not been found to surpass the non-biosynthetic NPs in antibacterial effects or vice versa.

Table 2 Antibacterial activities of green NPs

Immunomodulatory effects of nanomaterials based on metals

Metal-based NPs are known to trigger innate as well as adaptive immune responses.Citation86 The immunostimulatory potential of Ag, CeO2 and surface-modified Au NPs has been reported.Citation87 Immunostimulatory NPs may serve as double-edged swords by acting on bacteria as biocidal nanoweapons, as well as undermining the bacterial resistance to host immunity. ZnO NPs increased interferon gamma, TNF-α and IL-12 expression in primary human immune cells,Citation88 while in another study, IL-6, IL-1β, IL-8 and TNF-α were induced by ZnO NPs from peripheral blood mononuclear cells.Citation53 TiO2Citation89 and SiO2 NPsCitation90 activated inflammasomes and induced IL-1β release, which affected fibroblast proliferation. Ag NPs also induced inflammasome formation and triggered IL-1β release and subsequent caspase-1 activation.Citation91 Size-dependent immunomodulatory effect of Au NPs has been reported that may inhibit IL-12p70 production by DCs and Th2 polarization, or promote Th17 potentiation.Citation92 CeO2 NPs stimulated IL-10 production from DCs and triggered a strong Th2-biased cytokine profile. On the contrary, TiO2 NPs induced DCs to release IL-12 and polarize T cells to a Th1-bias.Citation93 TiO2 NPs and nanoplatinum triggered proinflammatory cytokine production, DC maturation and näive T cell activation and proliferation.Citation94,Citation95 A benign ε-polylysine/Ag nanocomposite has been reported to modulate the relative levels of CD3+ T cells and CD68+ macrophages and promote infected wound healing.Citation96

Synergistic antibacterial effects of nanomaterials with antibiotics or alternate antimicrobial compounds

While most metal-based NPs are microbicidal to an array of bacteria, genetic alterations in bacteria may result in rapid evolution of resistance to Ag NPs,Citation31 whereas Al2O3 NPs trigger increased expression of conjugation-promoting genes, thus promoting horizontal transfer of antibiotic resistance genes. NPs have been tailored to subdue resistance by packaging multifarious antimicrobial agents, resulting in development of resistance, which is a subtle possibility since it would require multiple simultaneous gene mutations in the same bacteria.Citation97,Citation98

The functionalization of NPs using antibiotics () is not only a promising nanoplatform to combat bacterial resistance but may also reduce the dose and hence toxicity of the drugs. NPs target or deliver antimicrobial agents to the infected site, thereby overcoming resistance as well as mitigating their hazardous impact on normal cells. Synergistic antibacterial efficiency of Ag NPs and antibiotics has been observed against S. aureus, E. coli and P. aeruginosa at extremely low concentrations.Citation99 The efficacy of ampicillin coupled with Ag NPs was identical in Gram-positive and Gram-negative bacteria, unlike the difficulty in inhibition of Gram-positive bacteria with Ag NPs alone.Citation39 Green synthesized NPs may also be used as an antibiotic adjuvant for the treatment of various bacterial infections (). Ag NPs boost the antimicrobial effects of several antibiotics, including penicillin G, amoxicillin, vancomycin, clindamycin and erythromycin against S. aureus, E. coli and MDR bacteria.Citation10 The antibiotic-functionalized NPs may promote reversal of antimicrobial resistance.

Table 3 Drug–nanomaterial synergy for antibacterial therapy

Combinatorial effect of Ag NPs with natural alternative compounds such as cinnamaldehydeCitation117 and eugenolCitation118 has also been reported. Se NPs conjugated with quercetin and acetylcholine had synergistic effect against MRSA, causing irreversible membrane damage.Citation119 Resveratrol nanocarriers with Au NPs and Ag NPs had potent activity against Gram-positive and Gram-negative bacteria.Citation120 The combination of biologically synthesized Ag NPs (produced by Fusarium oxysporum) and oregano (Origanum vulgare) essential oil showed bactericidal effects against non-methicillin-resistant S. aureus (non-MRSA) and β-lactamase- and carbapenemase-producing E. coli and A. baumannii strains.Citation121 Nigella sativa essential oil-synthesized Au NPs effectively inhibited the biofilm formation of S. aureus and Vibrio harveyi.Citation122

Polymeric nanocomposites and nanomaterials conjugated with antibodies and other ligands and drug–NP complex for targeted delivery

Antimicrobial NPs can be easily trapped within polymer films to form lipid–polymer hybrid NPs or nanocomposites. Enhanced biocompatibility and stability coupled with controlled release make these superior alternatives to metallic NPs. Synergistic antibacterial effects of some polymers and metal nanomaterials have been identified. The biocidal activities of Ag and Cu NPs embedded in polymer matrices have been reported to be enhanced due to release of metal ions while dampening their toxicities.Citation55,Citation123 Ag nanocomposites have been reported to be biocidal against MRSA.Citation124 A benign ε-polylysine/Ag nanocomposite demonstrated antibacterial effects against P. aeruginosa and S. aureus that was mediated by surface adherence, irreversible disruption of the membrane with subsequent penetration and inhibition of protein activity, ultimately leading to bacterial apoptosis.Citation96 Low-density polyethylene-containing ZnO nanocomposites have been found to be effective against B. subtilis and Enterobacter aerogenes.Citation125 Ag/Fe3O4 nanocomposites demonstrated high antibacterial activity against E. coliCitation126 and graphene-oxide Ag nanocomposites against MRSA.Citation127

Targeted NP delivery to the infection site could also be achieved by surface modification with ligands or antibodies, which may further improve therapeutic efficacy and reduce the side effects of antimicrobial drugs. The effectiveness of Ag NPs is also augmented with compounds such as polyethyleneimines, chitosan and glucosamine that serve as ligands and reinforce their uptake into bacterial cells.Citation128 Chitosan has inherent antimicrobial properties due its polycationic character. Low-molecular weight chitosan-coated Ag NPs have been reported to surpass polyvinylpyrrolidone-coated Ag NPs and Ag NPs without surface stabilizer (uncoated Ag NPs) in efficacy against MRSA with enhanced biocompatibility and reduced body absorption characteristics.Citation129 Chitosan/TiO2/Ag nanocomposites exhibited effective antibacterial activity via ROS generation, lactate dehydrogenase release and inhibited bacterial adhesion.Citation130 Chitosan/Ag nanocomposites were effective against Salmonella sp.,Citation131 whereas the activity of chitosan-TiO2:Cu nanocomposite against E. coli and S. aureus was enhanced in the presence of light.Citation132 Chitosan/calcium silicate nanocomposites doped with Ag+ exhibited antibacterial activity against S. aureus and P. aeruginosa.Citation133 Ag NPs capped with lipoic acid were effective against Staphylococcus epidermidis and Streptococcus mutans biofilm.Citation134 Ag-decorated poly (DL-lactide-co-glycolide) NPs exhibited high efficacy against S. epidermidis biofilms.Citation135 Biodegradable lignin-core NPs coated with cationic poly-electrolytes and infused with Ag+ exhibited broad-spectrum biocidal activity against Gram-positive as well as Gram-negative bacteria at lower concentrations of Ag+ than the conventional Ag NPs and hence suggested as greener alternatives to Ag NPs.Citation136 Antibacterial (P. aeruginosa) activity of iron oxide NPs was also enhanced upon chitosan coating.Citation137 Superior antibacterial activity of UV-irradiated glucosamine-functionalized Au NPs on graphene oxide against E. coli and Enterococcus faecalis has been reported that was better than kanamycin.Citation138 Gallic acid-capped Au NPs have been found to be effective against both Gram-positive and Gram-negative bacteria.Citation139 Synthetic peptides (containing arginine, tryptophan and cysteine termini)-immobilized Au NPs exhibited targeting capacity and activity against Staphylococci, Enterococci and antibiotic-resistant bacterial strains.Citation140 Microbial (Ochrobactrum rhizosphaerae) glycolipoprotein-capped Ag NPs exhibited antibacterial activity against V. cholerae comparable to ciprofloxacin.Citation141 Besides the targeting ligands, NPs conjugated with antibodies against surface antigen of the target microbe such as anti-protein A antibodies have been shown to have high selectivity for killing S. aureus. Conjugating Bi NPs to antibodies against the target has been found to be effective against MDR P. aeruginosa when irradiated with low-dose X-rays.Citation54 Similarly, the IgG in IgG-Fe3O4@TiO2 magnetic NPs has been shown to target S. pyogenes.Citation54

Drug–NP complex represents another type of polymeric NPs that are highly attractive as drug delivery vehicles due to their stability, ease of functionalization and sustained release. These may be made multifunctional by incorporating different polymers. Chitosan-functionalized Au NPs adsorbed on vancomycin-encapsulated liposomes released the antibiotic in the presence of anti-toxin antibody secreted by S. aureus, inhibiting its growth.Citation142 Lipids like phosphatidylinositol and stearylamine presented specific affinity with biofilms, thereby increasing the biofilm adhesion of liposomes. Anti-biofilm activities of phosphatidylcholine-decorated Au NPs loaded with gentamycin have been reported against P. aeruginosa, S. aureus as well as intracellular Listeria monocytogenes and E. coli.Citation143 NP-based drug delivery could subjugate systemic toxic effects of antibiotics, decrease uptake and increase efflux of drugs, biofilm formation and intracellular bacterial infection.

Conclusion and perspectives

Bacteria are increasingly dodging extermination as they have evolved innate immune resistance strategies. Antibiotics remain the mainstay to fight bacterial infections, but episodes of resistant infections are alarmingly on the rise. The indiscriminate use of antimicrobial agents in the community, hospital and agriculture is undeniably responsible for fuelling this crisis. This also causes selective pressure, allowing only the fittest genotype to thrive, resulting in emergence of MDR bacterial strains, and emphasizing the need for surrogate therapeutic options. The emergence of resistant and more virulent strains of bacteria has outpaced the development of new antibiotics over the last few decades. NPs are now being considered as a viable alternative to antibiotics due to their biocidal and immunopotentiating properties. The polymer-based nanomaterials and metal NPs may also be exploited as antimicrobial coatings on surface of medical devices for various biomedical applications. Metallic NPs when used with the existing antibiotics for bacterial infections lower the antibiotics dosage to be administered, thereby minimizing toxicity as well as reducing the probability of development of resistance. There has been a paradigm shift towards cost-effective and ecofriendly green synthesis of antibacterial NPs. This approach holds promise alone or synergistically with antibiotics targeting various bacterial infections, paving way for future therapeutics in nanomedicine. This combinatorial approach may serve as adjunct to the existing therapies and may help to restrain the escalating nosocomial threats. However, their translation to clinics would entail an in-depth understanding of the pharmacokinetics and biodistribution of NPs.

Disclosure

The author reports no conflicts of interest in this work.

References

  • Al-AnaziKAAl-JasserAMInfections caused by Acinetobacter baumannii in recipients of hematopoietic stem cell transplantationFront Oncol2014418625072028
  • SatoriusAESzafranskiJPyneDComplement c5a generation by staphylococcal biofilmsShock201339433634223459111
  • UbagaiTNakanoRNakanoAKamoshidaGOnoYGene expression analysis in human polymorphonuclear leukocytes stimulated by LPSs from nosocomial opportunistic pathogensInnate Immun201521880281226376669
  • FallahFAbdolghafoorianHSajadi NiaRSAntimicrobial resistance patterns of Acinetobacter baumannii, Pseudomonas aeruginosa and Staphylococcus aureus isolated from patients with nosocomial infections admitted to Tehran HospitalsArch Pediatr Infect Dis201534e32554
  • BeythNHouri-HaddadYDombAKhanWHazanRAlternative antimicrobial approach: nano-antimicrobial materialsEvid Based Complement Alternat Med2015201524601225861355
  • GuptaALandisRFRotelloVMNanoparticle-based antimicrobials: surface functionality is criticalF1000Res20165 F1000 Faculty Rev-364
  • PelgriftRYFriedmanAJNanotechnology as a therapeutic tool to combat microbial resistanceAdv Drug Deliv Rev20136513–141803181523892192
  • RaiMKonKGadeAAntibiotic resistance: can nanoparticles tackle the problem?KonKRaiMAntibiotic Resistance: Mechanisms and New Antimicrobial ApproachesAcademic Press, Elsevier IncUSA2016121
  • DijkshoornLNemecASeifertHAn increasing threat in hospitals: multidrug-resistant Acinetobacter baumanniiNat Rev Microbiol200751293995118007677
  • Japoni-NejadASofianMvan BelkumAGhaznavi-RadENosocomial outbreak of extensively and pan drug-resistant Acinetobacter baumannii in tertiary hospital in central part of IranJundishapur J Microbiol201368e9892
  • OmerMIGumaaSAHassanAAPrevalence and resistance profile of Acinetobacter baumannii clinical isolates from a private hospital in Khartoum, SudanAm J Microbiol Res2015327679
  • AljindanRBukharieHAlomarAAbdalhamidBPrevalence of digestive tract colonization of carbapenem-resistant Acinetobacter baumannii in hospitals in Saudi ArabiaJ Med Microbiol201564Pt 440040625657302
  • Cornejo-JuárezPVilar-CompteDPérez-JiménezCÑamendys-SilvaSASandoval-HernándezSVolkow-FernándezPThe impact of hospital-acquired infections with multidrug-resistant bacteria in an oncology intensive care unitInt J Infect Dis201531313425528484
  • VentolaCLThe antibiotic resistance crisis: part 1: causes and threatsPT2015404277283
  • LebeauxDGhigoJMBeloinCBiofilm-related infections: bridging the gap between clinical management and fundamental aspects of recalcitrance toward antibioticsMicrobiol Mol Biol Rev201478351054325184564
  • AnderssonDIHughesDKubicek-SutherlandJZMechanisms and consequences of bacterial resistance to antimicrobial peptidesDrug Resist Updat201626435727180309
  • SansonettiPJDi SantoJPDebugging how bacteria manipulate the immune responseImmunity200726214916117307704
  • GiedraitienėAVitkauskienėANaginienėRPavilonisAAntibiotic resistance mechanisms of clinically important bacteriaMedicina (Kaunas)201147313714621822035
  • MayersDAntimicrobial Drug Resistance: Volume 1. Mechanisms of Drug Resistance. Infectious DiseaseHumana PressUK2009
  • Alcalde-RicoMHernando-AmadoSBlancoPMartínezJLMultidrug efflux pumps at the crossroad between antibiotic resistance and bacterial virulenceFront Microbiol20167148327708632
  • ShaikhSFatimaJShakilSRizviSMKamalMAAntibiotic resistance and extended spectrum beta-lactamases: types, epidemiology and treatmentSaudi J Biol Sci20152219010125561890
  • BlairJMWebberMABaylayAJOgboluDOPiddockLJMolecular mechanisms of antibiotic resistanceNat Rev Microbiol2015131425125435309
  • CoxGStogiosPJSavchenkoAWrightGDStructural and molecular basis for resistance to aminoglycoside antibiotics by the adenylyltransferase ANT(2″)-laMBio201561
  • YunMKWuYLiZCatalysis and sulfa drug resistance in dihydropteroate synthase: crystal structures reveal the catalytic mechanism of DHPS and the structural basis of sulfa drug action and resistanceScience201233560721110111422383850
  • ZhuXRadovic-MorenoAFWuJLangerRShiJNanomedicine in the management of microbial infection – overview and perspectivesNano Today20149447849825267927
  • DizajSMLotfipourFBarzegar-JalaliMZarrintanMHAdibkiaKAntimicrobial activity of the metals and metal oxide nanoparticlesMater Sci Eng C Mater Biol Appl20144427828425280707
  • Hernandez-DelgadilloRVelasco-AriasDDiazDZerovalent bismuth nanoparticles inhibit Streptococcus mutans growth and formation of biofilmInt J Nanomedicine201272109211322619547
  • DakalTCKumarAMajumdarRSYadavVMechanistic basis of antimicrobial actions of silver nanoparticlesFront Microbiol20167183127899918
  • DuránNDuránMde JesusMBSeabraABFávaroWJNakazatoGSilver nanoparticles: a new view on mechanistic aspects on antimicrobial activityNanomedicine201612378979926724539
  • RaiMKDeshmukhSDIngleAPGadeAKSilver nanoparticles: the powerful nanoweapon against multidrug-resistant bacteriaJ Appl Microbiol2012112584185222324439
  • GravesJLJrTajkarimiMCunninghamQRapid evolution of silver nanoparticle resistance in Escherichia coliFront Genet201564225741363
  • AnsariMAKhanHMKhanAACameotraSSSaquibQMusarratJInteraction of Al(2)O(3) nanoparticles with Escherichia coli and their cell envelope biomoleculesJ Appl Microbiol2014116477278324354999
  • OktarFNYetmezMFicaiDFicaiADumitruFPicaAMolecular mechanism and targets of the antimicrobial activity of metal nanoparticlesCurr Top Med Chem201515161583158825877090
  • ŁysakowskaMECiebiada-AdamiecAKlimekLSienkiewiczMThe activity of silver nanoparticles (Axonnite) on clinical and environmental strains of Acinetobacter sppBurns201541236437125145873
  • SirelkhatimAMahmudSSeeniAReview on zinc oxide nanoparticles: antibacterial activity and toxicity mechanismNano-Micro Lett201573219242
  • WongMSChenCWHsiehCCHungSCSunDSChangHHAntibacterial property of Ag nanoparticle-impregnated N-doped titania films under visible lightSci Rep201551197826156001
  • ChatterjeeAKChakrabortyRBasuTMechanism of antibacterial activity of copper nanoparticlesNanotechnology2014251313510124584282
  • LelloucheJFriedmanALahmiRGedankenABaninEAntibiofilm surface functionalization of catheters by magnesium fluoride nanoparticlesInt J Nanomedicine201271175118822419866
  • FranciGFalangaAGaldieroSSilver nanoparticles as potential antibacterial agentsMolecules20152058856887425993417
  • YoonKYHoon ByeonJParkJHHwangJSusceptibility constants of Escherichia coli and Bacillus subtilis to silver and copper nanoparticlesSci Total Environ20073732–357257517173953
  • ShamailaSZafarNRiazSSharifRNazirJNaseemSGold nanoparticles: an efficient antimicrobial agent against enteric bacterial human pathogenNanomaterials (Basel)201664
  • LuoYHossainMWangCTargeted nanoparticles for enhanced X-ray radiation killing of multidrug-resistant bacteriaNanoscale20135268769423223782
  • HuoSJiangYGuptaAFully zwitterionic nanoparticle antimicrobial agents through tuning of core size and ligand structureACS Nano20161098732873727622756
  • KhashanKSSulaimanGMAbdul AmeerFANapolitanoGSynthesis, characterization and antibacterial activity of colloidal NiO nanoparticlesPak J Pharm Sci201629254154627087098
  • AliZRajBVishwasMAthharMASynthesis, characterization and antimicrobial activity of Ce doped TiO2 nanoparticlesInt J Curr Microbiol Appl Sci201654705712
  • NeethirajanSClondMAVogtAMedical biofilms–nanotechnology approachesJ Biomed Nanotechnol201410102806282725992419
  • LeeJHKimYGChoMHLeeJZnO nanoparticles inhibit Pseudomonas aeruginosa biofilm formation and virulence factor productionMicrobiol Res20141691288889624958247
  • LelloucheJFriedmanAGedankenABaninEAntibacterial and anti-biofilm properties of yttrium fluoride nanoparticlesInt J Nanomedicine201275611562423152681
  • GuisbiersGWangQKhachatryanEInhibition of E. coli and S. aureus with selenium nanoparticles synthesized by pulsed laser ablation in deionized waterInt J Nanomedicine2016113731373627563240
  • DhanabalanKGurunathanKMicroemulsion mediated synthesis and characterization of CdS nanoparticles and its anti-biofilm efficacy against Escherichia coli ATCC 25922J Nanosci Nanotechnol20151564200420426369030
  • LiakosIGrumezescuAMHolbanAMMagnetite nanostructures as novel strategies for anti-infectious therapyMolecules2014198127101272625140449
  • Nour El DinSEl-TayebTAAbou-AishaKEl-AziziMIn vitro and in vivo antimicrobial activity of combined therapy of silver nanoparticles and visible blue light against Pseudomonas aeruginosaInt J Nanomedicine2016111749175827175075
  • Esparza-GonzálezSCSánchez-ValdésSRamírez-BarrónSNEffects of different surface modifying agents on the cytotoxic and antimicrobial properties of ZnO nanoparticlesToxicol In Vitro20163713414127666655
  • GrumezescuANanobiomaterials in Antimicrobial Therapy: Applications of Nanobiomaterials1st edElsevierUSA2016
  • PerdikakiAGaleouAPilatosGAg and Cu monometallic and Ag/Cu bimetallic nanoparticle-graphene composites with enhanced antibacterial performanceACS Appl Mater Interfaces Epub20161011
  • AshfaqMVermaNKhanSCopper/zinc bimetal nanoparticles-dispersed carbon nanofibers: a novel potential antibiotic materialMater Sci Eng C Mater Biol Appl20165993894726652451
  • ParkYNew paradigm shift for the green synthesis of antibacterial silver nanoparticles utilizing plant extractsToxicol Res201430316917825343010
  • GandhiHKhanSBiological synthesis of silver nanoparticles and its antibacterial activityJ Nanomed Nanotechnol20167366
  • GhaediMYousefinejadMSafarpoorMKhafriHZPurkaitMKRosmarinus officinalis leaf extract mediated green synthesis of silver nanoparticles and investigation of its antimicrobial propertiesJ Ind Eng Chem201525167172
  • NayakDAsheSRautaPRKumariMNayakBBark extract mediated green synthesis of silver nanoparticles: evaluation of antimicrobial activity and antiproliferative response against osteosarcomaMater Sci Eng C Mater Biol Appl201658445226478285
  • AhmedMJMurtazaGMehmoodABhattiTMGreen synthesis of silver nanoparticles using leaves extract of Skimmia laureola: characterization and antibacterial activityMater Lett20151531013
  • SureshGGunasekarPHKokilaDGreen synthesis of silver nanoparticles using Delphinium denudatum root extract exhibits antibacterial and mosquito larvicidal activitiesSpectrochim Acta A Mol Biomol Spectrosc2014127616624632157
  • DuJSinghHYiTHAntibacterial, anti-biofilm and anticancer potentials of green synthesized silver nanoparticles using benzoin gum (Styrax benzoin) extractBioprocess Biosyst Eng201639121923193127495263
  • MengYSunYDevelopment of biogenic silver nanoparticle using Rosa chinensis flower extract and its antibacterial propertyJ Nanosci Nanotechnol20161643969397227451748
  • JunSHChaSHKimJHYoonMChoSParkYSilver nanoparticles synthesized using Caesalpinia sappan extract as potential novel nanoantibiotics against methicillin-resistant Staphylococcus aureusJ Nanosci Nanotechnol20151585543555226369115
  • DhandVSoumyaLBharadwajSChakraSBhattDSreedharBGreen synthesis of silver nanoparticles using Coffea arabica seed extract and its antibacterial activityMater Sci Eng C Mater Biol Appl201658364326478284
  • RameshMAnbuvannanMViruthagiriGGreen synthesis of ZnO nanoparticles using Solanum nigrum leaf extract and their antibacterial activitySpectrochim Acta A Mol Biomol Spectrosc2015136Pt B86487025459609
  • MaqboolQNazarMNazSAntimicrobial potential of green synthesized CeO2 nanoparticles from Olea europaea leaf extractInt J Nanomedicine2016115015502527785011
  • VenkateswarluSKumarBNPrathimaBAnithaKJyothiNVVA novel green synthesis of Fe3O4-Ag core shell recyclable nanoparticles using Vitis vinifera stem extract and its enhanced antibacterial performancePhysica B Condens Matter20154573035
  • TippayawatPPhromviyoNBoueroyPChompoosorAGreen synthesis of silver nanoparticles in aloe vera plant extract prepared by a hydrothermal method and their synergistic antibacterial activityPeerJ20164e258927781173
  • SinghKPanghalMKadyanSChaudharyUYadavJPGreen silver nanoparticles of Phyllanthus amarus: as an antibacterial agent against multi drug resistant clinical isolates of Pseudomonas aeruginosaJ Nanobiotechnology2014124025271044
  • MaparaNSharmaMShriramVBharadwajRMohiteKCKumarVAntimicrobial potentials of Helicteres isora silver nanoparticles against extensively drug-resistant (XDR) clinical isolates of Pseudomonas aeruginosaAppl Microbiol Biotechnol20159924106551066726362684
  • JangHLimSHChoiJSParkYAntibacterial properties of cetyltrimethylammonium bromide-stabilized green silver nanoparticles against methicillin-resistant Staphylococcus aureusArch Pharm Res201538101906191225893431
  • KrishnarajCJaganEGRajasekarSSelvakumarPKalaichelvanPTMohanNSynthesis of silver nanoparticles using Acalypha indica leaf extracts and its antibacterial activity against water borne pathogensColloids Surf B Biointerfaces2010761505619896347
  • RamalingamBParandhamanTDasSKAntibacterial effects of biosynthesized silver nanoparticles on surface ultrastructure and nanomechanical properties of Gram-Negative Bacteria viz. Escherichia coli and Pseudomonas aeruginosaACS Appl Mater Interfaces2016874963497626829373
  • MariselvamRRanjitsinghAJUsha Raja NanthiniAKalirajanKPadmalathaCMosae SelvakumarPGreen synthesis of silver nanoparticles from the extract of the inflorescence of Cocos nucifera (Family: Arecaceae) for enhanced antibacterial activitySpectrochim Acta A Mol Biomol Spectrosc201412953754124762541
  • SalemWLeitnerDRZinglFGAntibacterial activity of silver and zinc nanoparticles against Vibrio cholerae and enterotoxic Escherichia coliInt J Med Microbiol20153051859525466205
  • PatraJKBaekKHNovel green synthesis of gold nanoparticles using Citrullus lanatus rind and investigation of proteasome inhibitory activity, antibacterial, and antioxidant potentialInt J Nanomedicine2015107253726426664116
  • SalunkeGRGhoshSSantosh KumarRJRapid efficient synthesis and characterization of silver, gold, and bimetallic nanoparticles from the medicinal plant Plumbago zeylanica and their application in biofilm controlInt J Nanomedicine201492635265324920901
  • GopinathKKumaraguruSBhakyarajKGreen synthesis of silver, gold and silver/gold bimetallic nanoparticles using the Gloriosa superba leaf extract and their antibacterial and antibiofilm activitiesMicrob Pathog201610111127765621
  • PandianCJPalanivelRDhanasekaranSScreening antimicrobial activity of nickel nanoparticles synthesized using Ocimum sanctum leaf extractJ Nanoparticles201620164694367
  • AnsariMAKhanHMAlzohairyMAGreen synthesis of Al2O3 nanoparticles and their bactericidal potential against clinical isolates of multi-drug resistant Pseudomonas aeruginosaWorld J Microbiol Biotechnol201531115316425304025
  • AnandKTilokeCPhulukdareeABiosynthesis of palladium nanoparticles by using Moringa oleifera flower extract and their catalytic and biological propertiesJ Photochem Photobiol B2016165879527776261
  • KhirallaGMEl-DeebBAAntimicrobial and antibiofilm effects of selenium nanoparticles on some foodborne pathogensLWT – Food Sci Technol201563210011007
  • SonkusrePSingh CameotraSBiogenic selenium nanoparticles inhibit Staphylococcus aureus adherence on different surfacesColloids Surf B Biointerfaces20151361051105726590898
  • LuoYHChangLWLinPMetal-based nanoparticles and the immune system: activation, inflammation, and potential applicationsBiomed Res Int2015201514372026125021
  • JiaoQLiLMuQZhangQImmunomodulation of nanoparticles in nanomedicine applicationsBiomed Res Int2014201442602824949448
  • HanleyCThurberAHannaCPunnooseAZhangJWingettDGThe influences of cell type and ZnO nanoparticle size on immune cell cytotoxicity and cytokine inductionNanoscale Res Lett20094121409142020652105
  • YazdiASGuardaGRiteauNNanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1α and IL-1βProc Natl Acad Sci U S A201010745194491945420974980
  • PeetersPMPerkinsTNWoutersEFMossmanBTReynaertNLSilica induces NLRP3 inflammasome activation in human lung epithelial cellsPart Fibre Toxicol201310323402370
  • YangEJKimSKimJSChoiIHInflammasome formation and IL-1β release by human blood monocytes in response to silver nanoparticlesBiomaterials201233286858686722770526
  • TomićSĐokićJVasilijićSSize-dependent effects of gold nanoparticles uptake on maturation and antitumor functions of human dendritic cells in vitroPLoS One201495e9658424802102
  • SchanenBCDasSReillyCMImmunomodulation and T helper TH1/TH2 response polarization by CeO2 and TiO2 nanoparticlesPLoS One201385e6281623667525
  • SchanenBCKarakotiASSealSDrakeDR3rdWarrenWLSelfWTExposure to titanium dioxide nanomaterials provokes inflammation of an in vitro human immune constructACS Nano2009392523253219769402
  • GhoneumMGhoneumAGimzewskiJNanodiamond and nanoplatinum liquid, DPV576, activates human monocyte-derived dendritic cells in vitroAnticancer Res201030104075407921036722
  • DaiXGuoQZhaoYFunctional silver nanoparticle as a benign antimicrobial agent that eradicates antibiotic-resistant bacteria and promotes wound healingACS Appl Mater Interfaces2016839257982580727622986
  • BollenbachTAntimicrobial interactions: mechanisms and implications for drug discovery and resistance evolutionCurr Opin Microbiol2015271926042389
  • FischbachMACombination therapies for combating antimicrobial resistanceCurr Opin Microbiol201114551952321900036
  • PanáčekASmékalováMKilianováMStrong and nonspecific synergistic antibacterial efficiency of antibiotics combined with silver nanoparticles at very low concentrations showing no cytotoxic effectMolecules2015211E2626729075
  • NaqviSZKiranUAliMICombined efficacy of biologically synthesized silver nanoparticles and different antibiotics against multidrug-resistant bacteriaInt J Nanomedicine201383187319523986635
  • ThomasRNairAPKrSMathewJEkRAntibacterial activity and synergistic effect of biosynthesized AgNPs with antibiotics against multidrug-resistant biofilm-forming coagulase-negative Staphylococci isolated from clinical samplesAppl Biochem Biotechnol2014173244946024699812
  • HwangISHwangJHChoiHKimKJLeeDGSynergistic effects between silver nanoparticles and antibiotics and the mechanisms involvedJ Med Microbiol201261Pt 121719172622956753
  • WanGRuanLYinYYangTGeMChengXEffects of silver nanoparticles in combination with antibiotics on the resistant bacteria Acinetobacter baumanniiInt J Nanomedicine2016113789380027574420
  • BrownANSmithKSamuelsTALuJObareSOScottMENanoparticles functionalized with ampicillin destroy multiple-antibiotic-resistant isolates of Pseudomonas aeruginosa and Enterobacter aerogenes and methicillin-resistant Staphylococcus aureusAppl Environ Microbiol20127882768277422286985
  • IramSAkbar KhanJAmanNNadhmanAZulfiqarZArfat YameenMEnhancing the anti-enterococci activity of different antibiotics by combining with metal oxide nanoparticlesJundishapur J Microbiol201693e3130227226875
  • HurYEParkYVancomycin-functionalized gold and silver nanoparticles as an antibacterial nanoplatform against methicillin-resistant Staphylococcus aureusJ Nanosci Nanotechnol20161666393639927427725
  • KalhapureRSSonawaneSJSikwalDRSolid lipid nanoparticles of clotrimazole silver complex: an efficient nano antibacterial against Staphylococcus aureus and MRSAColloids Surf B Biointerfaces201513665165826492156
  • AkramFEEl-TayebTAbou-AishaKEl-AziziMA combination of silver nanoparticles and visible blue light enhances the antibacterial efficacy of ineffective antibiotics against methicillin-resistant Staphylococcus aureus (MRSA)Ann Clin Microbiol Antimicrob20161514827530257
  • GannimaniRRameshMMtamboSPillayKSolimanMEGovenderPγ-Cyclodextrin capped silver nanoparticles for molecular recognition and enhancement of antibacterial activity of chloramphenicolJ Inorg Biochem2016157152426824520
  • GuHHoPLTongEWangLXuBPresenting vancomycin on nanoparticles to enhance antimicrobial activitiesNano Lett20033912611263
  • PayneJNWaghwaniHKConnorMGNovel synthesis of kanamycin conjugated gold nanoparticles with potent antibacterial activityFront Microbiol2016760727330535
  • GhasemiFJalalRAntimicrobial action of zinc oxide nanoparticles in combination with ciprofloxacin and ceftazidime against multidrug-resistant Acinetobacter baumanniiJ Glob Antimicrob Resist2016611812227530853
  • GurunathanSBiologically synthesized silver nanoparticles enhances antibiotic activity against gram-negative bacteriaJ Ind Eng Chem201529217226
  • SinghRWaghPWadhwaniSSynthesis, optimization, and characterization of silver nanoparticles from Acinetobacter calcoaceticus and their enhanced antibacterial activity when combined with antibioticsInt J Nanomedicine201384277429024235826
  • HariNThomasNKNairAAJComparative study on the synergistic action of differentially synthesized silver nanoparticles with β-cephem antibiotics and chloramphenicolJ Nanosci20142014201482
  • KalitaSKandimallaRSharmaKKKatakiACDekaMKotokyJAmoxicillin functionalized gold nanoparticles reverts MRSA resistanceMater Sci Eng C Mater Biol Appl20166172072726838902
  • GhoshINPatilSDSharmaTKSrivastavaSKPathaniaRNavaniNKSynergistic action of cinnamaldehyde with silver nanoparticles against spore-forming bacteria: a case for judicious use of silver nanoparticles for antibacterial applicationsInt J Nanomedicine201384721473124376352
  • Perugini Biasi-GarbinRSaori OtaguiriEMoreyATEffect of eugenol against Streptococcus agalactiae and synergistic interaction with biologically produced silver nanoparticlesEvid Based Complement Alternat Med2015201586149725945115
  • HuangXChenXChenQYuQSunDLiuJInvestigation of functional selenium nanoparticles as potent antimicrobial agents against superbugsActa Biomater20163039740726518106
  • ParkSChaSHChoIAntibacterial nanocarriers of resveratrol with gold and silver nanoparticlesMater Sci Eng C Mater Biol Appl2016581160116926478416
  • ScandorieiroSde CamargoLCLancherosCASynergistic and additive effect of oregano essential oil and biological silver nanoparticles against multidrug-resistant bacterial strainsFront Microbiol2016776027242772
  • ManjuSMalaikozhundanBVijayakumarSAntibacterial, antibiofilm and cytotoxic effects of Nigella sativa essential oil coated gold nanoparticlesMicrob Pathog20169112913526703114
  • PalzaHAntimicrobial polymers with metal nanoparticlesInt J Mol Sci20151612099211625607734
  • ShuryginaIAShuryginMGSukhovBGNanobiocomposites of metals as antimicrobial agentsKonKRaiMAntibiotic Resistance: Mechanisms and New Antimicrobial ApproachesAcademic Press, Elsevier IncUSA2016167182
  • EsmailzadehHSangpourPShahrazFHejaziJKhaksarREffect of nanocomposite packaging containing ZnO on growth of Bacillus subtilis and Enterobacter aerogenesMater Sci Eng C Mater Biol Appl2016581058106326478403
  • GhaseminezhadSMShojaosadatiSAEvaluation of the antibacterial activity of Ag/Fe3O4 nanocomposites synthesized using starchCarbohydr Polym201614445446327083838
  • de MoraesACLimaBAde FariaAFBrocchiMAlvesOLGraphene oxide-silver nanocomposite as a promising biocidal agent against methicillin-resistant Staphylococcus aureusInt J Nanomedicine2015106847686126586946
  • AzevedoMMRamalhoPSilvaAPTeixeira-SantosRPina-VazCRodriguesAGPolyethyleneimine and polyethyleneimine-based nanoparticles: novel bacterial and yeast biofilm inhibitorsJ Med Microbiol201463Pt 91167117324913563
  • PengYSongCYangCGuoQYaoMLow molecular weight chitosan-coated silver nanoparticles are effective for the treatment of MRSA-infected woundsInt J Nanomedicine20171229530428115847
  • NatarajanSBhuvaneshwariMLakshmiDSMrudulaPChandrasekaranNMukherjeeAAntibacterial and antifouling activities of chitosan/TiO2/Ag NPs nanocomposite films against packaged drinking water bacterial isolatesEnviron Sci Pollut Res Int20162319195291954027388596
  • ArjunanNKumariHLSingaraveluCMKandasamyRKandasamyJPhysicochemical investigations of biogenic chitosan-silver nanocomposite as antimicrobial and anticancer agentInt J Biol Macromol201692778727381584
  • RautAVYadavHMGnanamaniAPushpavanamSPawarSHSynthesis and characterization of chitosan-TiO2:Cu nanocomposite and their enhanced antimicrobial activity with visible lightColloids Surf B Biointerfaces201614856657527693718
  • El-NahrawyAMAliAIAbou HammadABYoussefAMInfluences of Ag-NPs doping chitosan/calcium silicate nanocomposites for optical and antibacterial activityInt J Biol Macromol201693Pt A26727527543348
  • NiskaKKnapNKędziaAJaskiewiczMKamyszWInkielewicz-StepniakICapping agent-dependent toxicity and antimicrobial activity of silver nanoparticles: an in vitro study. Concerns about potential application in dental practiceInt J Med Sci2016131077278227766027
  • TakahashiCMatsubaraNAkachiYVisualization of silver-decorated poly (DL-lactide-co-glycolide) nanoparticles and their efficacy against Staphylococcus epidermidisMater Sci Eng C Mater Biol Appl20177214314928024570
  • RichterAPBrownJSBhartiBAn environmentally benign antimicrobial nanoparticle based on a silver-infused lignin coreNat Nanotechnol201510981782326167765
  • MukherjeeMDeSInactivation of Pseudomonas aeruginosa by chitosan coated iron oxide nanoparticlesRecent Pat Biotechnol201610113313927494734
  • GovindarajuSSamalMYunKSuperior antibacterial activity of GlcN-AuNP-GO by ultraviolet irradiationMater Sci Eng C Mater Biol Appl20166936637227612724
  • KimDYKimMShindeSSungJSGhodakeGCytotoxicity and antibacterial assessment of gallic acid capped gold nanoparticlesColloids Surf B Biointerfaces201714916216727756012
  • KuoYLWangSGWuCYFunctional gold nanoparticle-based antibacterial agents for nosocomial and antibiotic-resistant bacteriaNanomedicine (Lond)201611192497251027622499
  • GahlawatGShikhaSChaddhaBSChaudhuriSMMayilrajSChoudhuryARMicrobial glycolipoprotein-capped silver nanoparticles as emerging antibacterial agents against choleraMicrob Cell Fact2016152526829922
  • PornpattananangkulDZhangLOlsonSBacterial toxin-triggered drug release from gold nanoparticle-stabilized liposomes for the treatment of bacterial infectionJ Am Chem Soc2011133114132413921344925
  • MuHTangJLiuQSunCWangTDuanJPotent antibacterial nanoparticles against biofilm and intracellular bacteriaSci Rep201661887726728712