Int J Pharm Pharm Sci, Vol 13, Issue 8, 20-31Review Article

INORGANIC NANOPARTICLES: AN ALTERNATIVE THERAPY TO COMBAT DRUG RESISTANT INFECTIONS

MONIKA GUPTA1

1Department of Biotechnology, Invertis University, Bareilly, India, 1Amity Institutes of Biotechnology, Amity University Madhya Pradesh, Gwalior, India
Email: biotech.monikaphd@gmail.com

Received: 07 Mar 2021, Revised and Accepted: 09 Jun 2021


ABSTRACT

One of the most urgent challenges that medical sciences face today is overcoming the problem of drug resistance. This review paper encompasses research studies that provide a solution towards this major concern. It aims to highlight the therapeutic effects of various metal-based nanoparticles over conventional antibiotics. Severe infections caused by bacteria, viruses, fungi and parasites are transmitted easily and spread across millions of people round the globe. Resistance developed by these organisms and their various strains against regular antibiotics has posed great threat to save the lives of humans. Nanoparticles are tiny in nature and thus capable of generating Reactive Oxygen Species (ROS). These ROS bursts to create severe oxidative stresses causing damage to DNA, lipids peroxidation and protein changes resulting in cell death. This mechanism is quite different from traditional antibiotics and hence gives better results towards microbial resistance. The study demonstrates the use of metal nanoparticles such as silver, zinc oxide, aluminium oxide, gold, copper oxide, titanium dioxide, magnesium oxide, iron oxide in combination with various antibiotics to efficiently kill infectious microbes.

Keywords: Bacterial infection, Fungal infection, Viral infection, Parasite infection


INTRODUCTION

These infectious diseases spread through fungi, viruses, bacteria and parasites, which have become the cause of many deaths throughout the world [1]. Infectious diseases were classified 2 groups: emerging and re-emerging. New diseases were being referred as emerging infectious diseases whereas re-emerging diseases are not new but suffer from drug resistance and thus they again appear, due to which there is difficulty in treating and controlling them [2]. However, the ability to prevent infection of the human body is only possible by the immune system. Although some infections are direct, whereas others are very transmissible and dangerous [3]. Transmission of infections occurs when some microorganism enters the host cell, resulting in their replication inside the host cell, causing the tissue to be damaged. However, it is necessary to mention that some microbes can replicate the body's externally, which results in tissue damage [4].

Drug resistance largely interferes with the treatment of infectious diseases, which makes it necessary to develop a novel therapy which could overcome this resistance. The therapeutic agents such as any metal nanoparticles were used for the treatment of infectious diseases. Metal nanoparticles show antimicrobial activity, which depends on size. Small size of nanoparticles easily penetrates the bacterial cell wall, resulting in cell death [5]. Hence, drug delivery capability as well as their therapeutic efficacy is aimed at improvement at the pathological site. Metal nanoparticles have good properties [physico-chemical] [6]. The unique Physico-chemical properties of metal nanoparticles make them a potent weapon against infectious diseases. Hence, these were designed for the majority of biomedical applications [7]. This review focuses on the biological activity of metal-based nanoparticles that are affordable and non-toxic and provide potential therapeutics against various fungal, parasitic, viral and bacterial diseases.

Search criteria

Exploring the latest knowledge over the subject through Research gate, Pubmed, EMBASE, Google scholar, Google web searches, SCOPUS and Web of Science greatly assisted in writing this review paper. Updated information has been incorporated on the basis of an extensive literature search on this subject.

Mechanism of nanoparticles to microbes

As a supplement to antibiotics, Nanomaterials as antimicrobials are highly promising and are receiving great interest as they can fill the gap where antibiotics are often unsuccessful. It includes combatting multidrug-resistant biofilm and mutants [8, 9]. An antibacterial nanoparticle is now in use [metal, metal oxide, and organic NPs] indicates the diversity of internal and modified chemical composition properties. Thus, it is unexpected that they have several ways [fig. 1]. Apart from this, there is considerable variation in the genetics of the target bacteria, as well as inhibiting them in cell wall structure, essential metabolic pathways and many components can prove to be very fatal for microorganisms. In addition, the physiological condition of bacteria, i.e., the planktonic, growth rate, biofilm, the sensitivity of the bacteria to stable or starved, can contribute significantly to the nanomaterials [10, 11]. In some cases, the ratio between bacteria and nanomaterials is important for subsequent toxicity. Furthermore, many environmental factors play a role and affect nanomaterials lethality, which contain bacteria, including pH, aeration and temperature [12]. With other nanoparticles, the physiological properties of particles including chemical modification, coating, size, shapes, and mixing in different ratios and the use of solvent affect all their antibacterial activity [13]. Thus, the Mode of Action and Level of hazard of Nanomaterials Antibacterial is still unclear and literature can find in other reports [14, 15]. However, as usually, nanomaterials works with two major routes, Which are related to each other and occur in many cases simultaneously: 1. reactive oxygen The production of species [ROS], also known as oxygen-free radicals, acts as nanomaterials nanocatalysts and with different chemical structures to generate ROS associated with their hazardous and toxic effects has been well-characterized in previous studies [16, 17]. Compared with microparticles or their bulk of origin, NPs possess unique physicochemical properties [size, surface area, shape, solubility, and aggregation status] that correlate with their potential to generate ROS [18-24]. 2. Disruption of membrane potential and integrity [25, 26].

Membrane damage occurs when the nm tie electrostatic bacteria goes to the cell wall and membrane, causing loss of membrane potential, membrane depolarization, and integrity, which in turn, imbalance of transport, impaired respiration, obstruction of energy transit And/or cell analysis, and ultimately cell death [27].

The most effective determinant for both in vitro and in vivo cytotoxicity of ROS, Nanomaterials is believed to be, by the disruption of the respiratory chain itself or directly induced by the Nanomaterials [28]. A burst of ROS occurs through severe oxidative stress, causing damage to all the brain cells, thereby prohibiting lipid peroxidation, protein changes, enzymes and RNA and DNA damage. In high doses, ROS leads to cell death and low dose, causing severe DNA damage and mutations [29, 30]. In some cases, where ROS production is visible or induced by UV light [31] NM poisoning is photocatalytic, for example as show in table 1, TiO2 nm was induced under almost-UV light, lipid peroxidation, which was E. coli leads to respiratory disease and death of cells [32]. Several other effects of nanomaterials include direct inhibition of specific essential enzymes, induction of nitrogen reactive species [NRS] [30, 12, 15, 16] and induction of programmed cell death [31].

Fig. 1: Mode of action of nanoparticles on bacteria (Adapted and modified)

Table 1: Various nanomaterials kill to various bacterial cell wall

Nanoparticles Size [nm] Microbes Time Concentration Target site Reference
Ag 9.3 E. coli 10 min
Cell wall [33]
Au 25 C. pseudotuberculosis 20 min 50, 100 and 200 µg/ml Cell wall [34]
Fe3O4 Ag 60 E. coli, S. epidermidis, Bacillus subtilis 24 h 60-70 μg/ml Cell wall [35]
MgO 4 E. coli, B. megaterium 20-60 min
Cell wall [36]
ZnO 30 Salmonella enteric, Escherichia coli 16 h 0.5 mg/ml Cell wall [37]
Cu 100 B. subtilis, E. coli 24 h 60 μg/ml
[38]
TiO2 8 Staphylococcus aureus 30 min - Cell wall [39]
Al2O3 60 E. coli, B. subtilis, Pseudomonas - 20 μg/ml Flocculation [40]

Classification of infections

Infectious disease is a clinically clear disorder that is caused by the presence of a pathogenic agent that can be either a bacteria, fungus, virus or parasite, from these diseases to one person [tuberculosis, malaria,] and due to the ability to move from one species to another [influenza, flu] sometimes called these communicable diseases. Infectious diseases can be highly classified: 1] known diseases which are insistent [e. g., tuberculosis, dengue, malaria,]; 2] New, previously unknown diseases [e. g., severe acute respiratory syndrome]; And 3] threatening to progress in the near future [e. g., avian influenza] is a major threat to these diseases because more than half of the world's deaths occur in these diseases, especially in developing countries [41]. Parasitism causes the benefits and infections received by pathogenic bacteria attacking the host [42]. These infections are described in below.

[i] Bacterial infections treated with metal-based nanoparticles

Treatment of bacterial infection is being interrupted by drug resistance due to which the danger is increasing on all over the world. Several molecular strategies have also been developed which increase their adhesion for host cells and their ability also gives the result of their colonies [43]. One such molecular strategy is hair-like organelles which are known as pili on the surface of bacteria. Bacteria are bound with host cells through these pili. Only Gram-positive and Gram-negative bacteria possess hair-like organelles structure [43-45]. Another probable molecular strategy is through forming biofilms that would protect bacteria in adverse conditions [43, 46]. Biofilm formation consists of protein, exopolysaccharides, EDNA etc. and has three main steps, i.e. the attachment to the surface of the host cell, the formation and separation of biofilm structure [47]. These components resists antibiotics and antibacterial agents. Biofilm creates adhesion between bacterial cells, from which multi-layered biofilms are formed. This segment will focus on the antibacterial activity of metal-based antibacterial compounds.

Silver nanoparticles

Several papers have been reported on silver nanoparticles penetrated to the bacterial cell wall. Subsequently, damage the structure of cell wall then cell death as shown in [fig. 1]. Some researchers have proposed that the silver nanoparticles produce free radicals and interact with bacteria and release silver ion, which causes cellular enzyme deactivation and inhibits several functions in cell and causing cell death. Another study reported that silver nanoparticles interact with DNA, inhibit to bacterial DNA replication, resulting cell death [70-77]. Tuberculosis is an infectious disease that affects the lungs and bacterium are caused by Mycobacterium tuberculosis [48-50]. Silver nanoparticles inhibited the growth of Mycobacterium tuberculosis in vitro and showed low cytotoxic effects. Silver nanoparticles were selected for the treatments and their therapeutic outcome as shown in (table 2).

Table 2: Various metal-based nanoparticles with antimicrobial activity their therapeutic outcome

Metal nanoparticles Therapeutic effect References
AgNPs Hampers growth of Mycobacterium tuberculosis [48-50]
AgNPs in urinary catheter Potent against urinary tract infections causing bacteria [51-56]
AgNPs combined with blue light and either of the Clarithromycin, Azithromycin, Amoxicillin, Linezolid Or Vancomycin Very efficient against Staphylococcus aureus. [57]
AgNPs synthesised by plants extracts Stable and antibacterial nanoparticles. [58-69]
AgNPs isolated from industrialized area Molecular identification Bacillus strain CS 11 [70]
AgNPs Effective against Staphylococcus aureus and Escherichia coli [71-77]
AgNPs generated using biological Methods [virus, fungi etc.] Efficient antibacterial activity. [78-84]
AgNPs+rifampicin and polymixin B Very Effectively treat Acinetobacter baumannii infection. [85]
AgNPs+amoxicillin Effectively treat Escherichia coli [86]
AgNPs prevention of gastrointestinal [87]
Al2O3NPs Good Antimicrobial sensitivity against Escherichia coli [88]
Al2O3NPs Nanoparticles entered Candida cells to disrupt their physiological activity. [89-90]
Al2O3NPs prepare using plant extract Effectively treat P. aeruginosa [91]
Al2O3NPs Potent against gram-negative and gram-positive bacteria [92, 93]
AuNPs combined with ofloxacin Superior bactericidal property [94]
AuNPs combined with gentamicin Effective against Escherichia coli [95, 96]
AuNPs combined with kanamycin, ampicillin, streptomycin and levofloxacin High potency against Micrococcus luteus, E. coli, and Staphylococcus aureus [97, 98]
AuNPs Highly active against pathogens and multi-drug resistant, Gram-negative and Gram-positive pathogens [99]
AuNPs prepare using Mulberry leaf extract

Effective against human pathogen

Vibrio cholera [gram-negative] and Staphylococcus aureus [gram-positive]

[100]
Cu2O NPs Good antibacterial activity against Bacillus sp. FU4 [101]
Cu2O NPs prepare using plant extract Good antibacterial activity against S. dysenteriae, Vibrio cholerae non.0139 [L4], Vibrio cholerae non.0139 [CSK6669], S. pneumoniae, S. aureus and E. coli [102-104]
Cu2O NPs Highly active [105]
Cu2O NPs

Antibacterial against Pseudomonas fluorescens, Aeromonas

Hydrophila and Flavobacterium branchiophilum,

[106]
FeNPs prepare using plant extract Antibacterial properties Escherichia Coli, Pseudomonas Aeruginosa, and Staphylococcus Aureus [107, 108]
Fe2O3NPs+erythromycin Effective against S. pneumonia [109]
Fe2O3NPs combined with ciprofloxacin Poor antibacterial activity [110]
Ga NPs Hampered mycobacteria [111]
Ga NPs Effective against Pseudomonas aeruginosa [112]
Ga NPs Strongly inhibited Mycobacterium tuberculosis [113]
Ga NPs Destroyed metabolism of F. Tularensis Fe [114]
MgONPs using plant extract of Swertia chirayaita Gram+ve bacteria S. epidermidis, S. aureus, B. cereus and Gram-ve bacteria, P. vulgaris, E. coli, K. pneumonia [115]
Tio2 NPs Effective against Streptococcus mutans and E. coli [116-121]
Tio2 NPs Potent against S. aureus [122]
ZnONPs nanoparticles coated wit gentamicin Significant antibacterial activity against Pseudomonas aeruginosa, Escherichia coli, Staphylococcus aureus, Listeria monocytogenes, Bacillus cereus [123]
ZnONPs prepare using Azadirachta indica Good antimicrobial activity against Gram-positive and Gram-negative bacteria: S. aureus, S. pyogenes and Escherichia coli [124]
ZnONPs Concentration of the nanoparticles determined inhibition of B. subtilis [125, 126]

Furthermore, silver nanoparticles also treat urinary tract infections which are caused by P. aeruginosa and Enterobacter.

These were also used in synergy with antibiotic to treat bacterial infection thereby enhancing the therapeutic outcome. Li et al. suggested the combination of amoxicillin and silver nanoparticles against Escherichia coli [85] to check for selected functionalities [hydroxyl and amido-groups] on amoxicillin with silver nanoparticles, which resulted in the formulation of powerful antibacterial activity. Apart from this, Wan et al. reported that the synergistic effect aginst Acinetobacter baumannii to combine the antibiotics polymixin B and rifampicin with the silver nanoparticles, which are treated with hospital-acquired infections [86]. The possible mechanism for this synergistic effect was due to the disruption of the bacterial cell wall by Ag+ion thereby producing cytotoxic ROS by the visible blue light.

Silver nanoparticles have been prepared using plant extract which contains secondary metabolites such as vincristine, tannin, flavanoids and polyphenols which helps to form Ag nanoparticles. Examples include Catharanthus roseus [C. roseus] [L.] G. Don,
[13], neem leaves [12], aloe vera [14], Parkia speciosa Hassk pods [11]. These plant extracts were used as reducing and stabilizing agents and show antibacterial potency [58-69].

Aluminium oxide nanoparticles

Aluminium Oxide nanoparticles exhibit antimicrobial activity against pathogenic microbes. An Aluminium Oxide nanoparticle penetrates to bacterial cell wall, resulting damage to all respiratory function and cell death [fig. 1]. Ansari et al. reported the green synthesis of Aluminium Oxide nanoparticles using leaf extract of lemon grass and their antimicrobial activity against extended-spectrum metallobeta-lactamases and beta-lactamases of clinical isolates of P. aeruginosa was found greater [91], Jalal et al. reported that Aluminium Oxide nanoparticles generation using leaf extract of Cymbopogon citratus. These nanoparticles get attached to the surface of bacterial cell wall, disrupting physiological activity and causing cell death [89, 90]. Sadiq et al. Reported that Aluminium Oxide nanoparticles are demonstrated antimicrobial activity against Escherichia coli [87, 88, 92, 93].

Gold nanoparticles

Ahamad et al. reported that gold nanoparticles combine with antibiotic ofloxacin, resulting exhibit superior antimicrobial activity [94]. Other researches have also reported that the gold nanoparticles when combined with antibiotics gentamicin are efficient against both Gram-positive and Gram-negative bacteria [95, 96]. The combine antibiotic with gold nanoparticles to enhanced the antimicrobial activity. In addition, Saha et al. reported that the combines kanamycin, ampicillin, streptomycin and levofloxacin [97, 98]. With the gold nanoparticles, which are treated with E. coli, Micrococcus luteus and Staphylococcus aureus infections [95-98]. Conjugating the antibiotic to the nanoparticles resulted enhanced antimicrobial efficiency when compare to antibiotic alone suggesting that the gold nanoparticles enhance interaction with the bacterial cell wall, resulting in cell death. Furthermore, Advallane et al. was reported that biosynthesis of gold nanoparticles using leaf extract of mulberry against human pathogen Vibrio cholera [gram-negative] and Staphylococcus aureus [gram-positive].

Copper oxide nanoparticles

Taran et al., showed that Copper oxide nanoparticles demonstrated antimicrobial activity against bacterial strains and are highly active against Bacillus sp. FU4 [101]. Chatterjee et al. have reported similar finding in which the antibacterial activity of copper oxide is due to generation of ROS nand DNA degradation in bacterial cells as show in fig. 1 [105]. The copper oxide nanoparticles are attributed to attach to surface of the bacterial cell, disrupted cell wall and causing cell death. The antimicrobial activity is depends on the particles size. The small size of nanoparticles shows high antibacterial activity. The spherical shapes of copper oxide nanoparticles show the highly antimicrobial activity against Aeromonas hydrophila, Pseudomonas fluorescens and Flavobacterium branchiophilum [106].

Iron oxide nanoparticles

Massadeh et al., investigated that iron oxide nanoparticles combine with ciprofiloxin, exhibited the poor antibacterial activity which may be due to interaction with ciprofiloxin inhibiting the absorbance of nanoparticles on the surface of bacterial cell [110]. But Aparicio-Caamaño et al. suggested that the combination that could inhibit S. pneumonia growth: Fe nanoparticles with erythromycin. This was due to enhanced entry of erythromycin into the bacterial cell wall due to the FeO nanoparticles [100]. Rafi et al. reported that plant extract could be used to produce iron oxide nanoparticles where concentration would determine the shape,size and antimicrobial activity of the nanoparticles [98, 99].

Gallium nanoparticles

Gallium nanoparticles also exhibited antibacterial activity [table 2]. Narayanasamy et al., investigated that GaNPs hampered the growth of mycobacterium for 15 d after a single drug-loaded [111]. Kurtjak et al., investigated that Gallium nanoparticles with the good antimicrobial activity against p. aeruginosa [112]. Choi et al., suggested that the prepare of gallium nanoparticles for enhanced the antimicrobial activity against Mycobacterium tuberculosis infection [113]. The nanoparticles enhanced the maturation of phagosome, which indicates their potential as anti-tuberculosis drugs [114].

Magnesium oxide nanoparticles

Magnesium oxide nanoparticles are attributed to the attachment to the surface of the bacteria cell wall, distrusting the ATP, resulting the cell death. Gaurav et al., Magnesium oxide nanoparticles using plant extract and show antimicrobial activity against Gram+ve bacteria [S. aureus, S. epidermidis, B. cereus] and Gram–ve bacteria [E. coli, P. vulgaris, K. pneumonia]. There are few paper published [106].

Titanium dioxide nanoparticles

Titanium dioxide nanoparticles’ antimicrobial activity is due to interaction with surface of bacterial cell where photo-catalytic action results in cell permeability and therefore cell death. Lin et al., prepared the Titanium dioxide nanoparticles with smaller size which produces the high content of ROS. The smaller surface area resulted in greater penetration and thus more damage of membrane [116-121].

Zinc oxide nanoparticles

Raghupati et al., prepared smaller zinc oxide nanoparticles which resulted in nanoparticles with good antibacterial activity. Hsueh et al., reported the inhibition effect of ZnO NPs on the growth of B. subtilis to depend solely on nanoparticles concentration. The accumulation of nanoparticles on the outer membrane of bacterial cell resulted cell death [125-126]. Voicu et al., coated ZnO NPs with gentamicin and determined their antimicrobial activity against, Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, Listeria monocytogenes and Bacillus cereus [123].

[ii] Fungal infections treated with metal-based Nanoparticles

Fungus is a ubiquitous and diverse group of micro-organisms. Some groups of fungi are plant pathogens which can cause serious infections in human beings [127, 128]. Skin disorders are due to a fungal group known as dermatophytes [129, 130]. Dermatophytoses may lead to major problems in immune-compromised hosts, and growing expansion of fungal skin infections in patients with HIV [131, 132]. In addition, prophylaxis with Antifungal may be due to the emergence of resistant strains [133]. Therefore, there is a need to search for a new generation of antifungal agents [134, 135].

Silver nanoparticles

Only few studies have reported the effects of silver nanoparticles on dermatophytes as most of them focused on the effect on Candida species [136-143]. The antifungal activity of AgNPs in biostabilized footwear materials was evaluated against dermatophytes and other fungi [144]. Noorbakhsh et al. [145, 146] investigated the effects of biologically synthesized silver nanoparticles by Klebsiella pneumoniae against Trichophyton rubrum.

Copper oxide nanoparticles

Sengal et al. found the efficacy of Copper oxide nanoparticles combined with fluconazole against Candida albicans infection [147]. Gold nanoparticles are potential anti-fungal agents which are developed by plasmonic clinical sample synthesis. The gold nanoparticles were negatively charged and inhibited the cell, replication and good antifungal effect [148].

Magnetic nanoparticles have also been found to be effective against the antifungal effect. Niemirowicz et al., suggested antifungal activity demonstrated against C. glabrata, C. tropicalis, and C. albicans [149, 150].

Titanium dioxide nanoparticles

The antifungal activity of titanium dioxide nanoparticles attributes to interaction with cell surface of fungi where photo-catalytic action results in cell permeability and cell death. Haghighi et al., prepared the Titanium dioxide nanoparticles with smaller size which produces the high content of ROS. The smaller surface area resulted in greater damage of the membrane [151].

Zinc oxide nanoparticles

Sardella et al., prepared zinc oxide nanoparticles which demonstrated good antifungal activity against Penicillium expansum and Botrytis cinerea [152, 153]. Xue et al., reported that zinc oxide nanoparticles inhibit growth of fungal by the accumulation of nanoparticles on the outer membrane of the fungal cell causing cell death [154]. In addition, it also showed its antifungal activity against A. flavus and A. fumigates [155, 156]. Grijaba et al. reported that the zinc oxide nanoparticles with antifungal activity against Erythricium salmonicolor [157].

Viral infections treated with metal-based nanoparticles

Some viral infections are persistent and cannot be removed from the body through the immune system alone such as Herpes, Hepatitis, HIV, etc. which may occur for many years. There are reports that metal-based therapeutics are effective to treat viral infections as explained below.

HIV

HIV is a viral infection with 36.7 million people suffering from HIV/AIDS worldwide [158-162].

Lara et al., reported the use of silver nanoparticles as an anti-HIV activity in both the initial phase of viral multiplication and entry-level of the HIV-1 life cycle [158-162]. In the initial phase, the silver nanoparticles got bind to gp120, as it inhibits the CD4-dependent virion binding, its fusion and also its infectivity. With the use of gp120 glycoprotein, silver nanoparticles prevent the virus from binding on host cells [162].

Kesarkar et al. reported that coated gold nanoparticles with polyethylene glycol exhibited greater antiviral activity. Kesarkar et al. further reported that the most effective concentration of gold nanoparticles at 2 µg/g and 4 µg/g were used for inhibiting virus entry [174].

Table 3: Various metal-based nanoparticles with antifungal potency and their therapeutic effect

Metal nanoparticles Therapeutic effect References
Silver nanoparticles prepare using Erythrina suberosa Antifungal Activity against C. albicans, C. kruseii, T. mentagrophytes, and C. viswanathii. [136]
silver nanoparticles biofilms Antifungal Activity against Candida albicans [137]
Silver nanoparticles Antifungal Activity against Candida species [138-142]
Amphotericin B-conjugated biogenic silver nanoparticles Antifungal activity against Candida albicans and Candida tropicalis [143]
Silver nanoparticles Antifungal Activity against Foot mycosis prophylaxis [144]
Silver nanoparticles Antifungal Activity against Trichophyton rubrum [145, 146]
Copper oxide nanoparticles combine with fluconazole Antifungal activity against Candida albicans [147]
Gold nanoparticles Human Cutaneous Preventing From fungal infection [148]
Magnetic nanoparticles Candidacidal activity against C. albicans, C. glabrata and C. tropicalis [149]
Iron-oxide nanoparticles Antifungal effect against different Candida species [150]
TiO2 nanoparticles Prevention of fungal biofilms especially biofilms formed on the surface of medical devices. [151]
Zinc oxide nanoparticles Antifungal activity against Penicillium expansum and Botrytis cinerea [152, 153]
ZnO nanoparticles Inhibit the fungal growth and benefit to public health and environment [154]
ZnO-NPs Antifungal Activity against A. flavus and A. fumigates [155, 156]
ZnO-NPs Antifungal activity against Erythricium salmonicolor [157]

Table 4: Various metal-based nanoparticles and their antiviral efficacy with therapeutic results

Metal nanoparticles Type of infection Therapeutic result References
Ag nanoparticles HIV Inhibits CD4-dependent viral binding. [158-162]
Ag nanoparticles Herpes Prevention virus infection [163, 164]
Ag nanoparticles Hepatitis Inhibits the production of HBV RNA and extracellular virions by interacting with HBV viral particles [165-167]
Ag nanoparticles Influenza Damages morphological structure of influenza virus. Prevents binding sites of the virus. [168-173]
Au [Gold] nanoparticles HIV Inhibits entry of virus [174]
Au nanoparticles Herpes Inhibited attachment of virus and stops penetration into the cell. [175, 176]
Au nanoparticle combined with interferon-alpha Hepatitis Targets interferon alpha [177, 178]
Au nanoparticles Influenza Very reliable against influenza A virus [179, 180]
FeO nanoparticles Hepatitis Hampered hepatitis C virus gene, protease and helicase, NS3. HCV NS3 gene encodes. [181]
Cuprous nanoparticle Hepatitis Prevented virus entry that involved genotypes [1a, 1b, and 2a] thus restricting viral replication [182]
ZnO nanoparticle Herpes Prevented viral entry and infection [183, 184]

Herpes

Herpes simplex virus HSV-1 and HSV-2 causes Herpes.

When Silver nanoparticles and HSV-2 interacts, it results in the reduction of offspring of the virus with weak cytotoxicity in vitro [163]. The High concentrations of silver nanoparticles are toxic to Vero cells. 100 µg/ml concentration of silver nanoparticles was used for inhibit the viral replication.

Baram-Pinto et al., reported the inhibition of HSV-1 virus by coating gold nanoparticles with mercarptoethene sulfonate. When the gold nanoparticles get attached to the surface of cell membrane, it inhibits viral entry and prevents from infection. These gold nanoparticles are non-toxic and very much useful for the treatment of viral infection [175, 176].

Mishra et al., developed micro nanoparticles of zinc oxide nanoparticles coated with various nanoscopic spikes that mimicks cell induced filopodia, which bind to their cell surface and inhibit viral entry and prevent infection [183, 184].

Hepatitis

Hepatitis is a viral infection which are of various types namely A, B, C, D, E and G type Hepatitis. Hepatitis A is caused by RNA virus that spreads through oral route and sexual contact [174]. Hepatitis B is caused by DNA virus that pass on genetically and sexually. Hepatitis C is caused by RNA virus and passes on by infected blood or sexually. In addition, Hepatitis D is caused by RNA virus. It can be found only in those who are infected with Hepatitis B virus [HBV]. The dual infection of Hepatitis B virus [HBV] and Hepatitis D virus [HDV] can result in more serious disease and worse outcome. Hepatitis E virus is transmitted through contaminated water or food.

Hepatitis G virus is recently discovered and belongs to Flaviviridae family. Silver, gold, iron oxide and cuprous oxide nanoparticles were used for the treatment of hepatitis infections [table 4].

Lu et al., investigated the nanosize [10 nm and 50 nm] of silver nanoparticles were studied in vitro as anti-HBV on the HepAD38 cell line. It has been observed that nanoparticles reduced the external HBV DNA formation of hepAD38 cells by 50%. These silver nanoparticles inhibits the production of hepatitis B virus RNA and virions [165-167].

Yarshi et al., developed In vitro evaluation on a blood sample infected with hepatitis C virus for hepatitis C virus viral loaded in a 1:1 ratio and gold nanoparticles didn’t show any activity on the virus [177, 178]. But they act as drug delivery systems such as Hyaluronic acid-gold nanoparticles combination was used to treat hepatitis C infection to the delivery of interferon-alpha in liver. Ryoo et al., investigated that iron oxide nanoparticles were inhibited to hepatitis C virus gene, NS3 and encoded helicase and protease which are responsible for viral replication.

Similarly, 2µg/ml concentration of cuprous oxide nanoparticles was used for inhibition of viral replication. These cuprous oxide nanoparticles are attached to the surface of cell membrane to inhibit viral entry, to prevent from infection of hepatitis C [182].

Influenza

Influenza is a viral infection that spreads through the respiratory system which is spread all over the world, especially among people suffering from chronic diseases. Xiang et al., reported the treatment of H3N2 influenza viruses by silver nanoparticles as they damage the morphological structure of the virus [168-173]. The damage was time-dependent. Li et al., investigated the delivery of silver nanoparticles in combination with amantadine to inhibit the H1N1 infection by ROS accumulation [168-173]. Other reports on silver nanoparticles also suggest about to inhibit the influenza virus and show anti-influenza activity, respectively [168-173]. Thus the antiviral activity was due to preventing virus attachment to the cell surface.

Parasitic infections

There are many examples of parasitic infections such as malaria, leishmaniasis, helminths etc. which suffer from drug resistance due to poor treatment compliance.

Malaria

Malaria is a mosquito-borne infection with 210 million people suffering from Malaria. In 2015, it was estimated that there were 438,000 people died in all over world [185-186]. It is spread by mosquitoes and caused by a parasite protozoan belonging to the Plasmodium type. The major challenge to treat malaria infections is drug resistance, which is due to bad treatment, mutation rates of parasites, efficacy of selected medicines and various strains of malaria parasites, which are responsible for co-infection.

Silver nanoparticles

There are few reports on silver nanoparticles against the antiplasmodial activity. Mishra et al., reported that growth of P. falciparum in human R. B. C culture was inhibited by silver nanoparticles synthesized using leaf extract. These biosynthesized silver nanoparticles were used against antiplasmodial activity at concentration IC50 [g/ml] of 3.75 for Amylase [185]. Murugan et al., reported the silver nanoparticles to have shown antiplasmodial activity against chloroquine-sensitive and chloroquine-resistant strains of Plasmodium falciparum at [IC50] 72.45 and 76.08 µg/ml, respectively when using C. tomentosum as a reducing, capping and stabilizing agent [187].

Selvam et al., developed biosynthesized silver nanoparticles using Catharanthus roseus leaf extract as a reducing and stabilizing agents, which showed antiplasmodial activity against P. falciparum at 25, 50, 75, and 100 µg/ml, respectively [189].

Gold nanoparticles

Dutta et al., reported that gold nanoparticles synthesized using bark extract of Syzygium jambos, demonstrated antiplasmodial activity against chloroquine-resistant strains of P. falciparum at 51.63 and 49.38 µgm/l while activity against chloroquine-sensitive strains of P. falciparum at IC50 values of 49.54 and 45.49 µg/ml [202, 203].

Leishmaniasis

Leishmaniasis is caused by Leishmania parasites. It is caused by the bite of infected female phlebotomine sandflies. Over 90 sandfly are known to transmit Leishmania parasites. Some of the anti-leishmanial drugs suffer from drug resistance [197, 198] Some researches have revealed treatment of leishmanial infection by metal-based nanoparticles [table 5].

Silver nanoparticles

Lima et al., found out an antileishmanial activity against promastigote forms of Leishmania amazonensis by synthesizing chitosan-based silver nanoparticles which demonstrated antileishmanial activity at MIC ranging from 1.69 to 3.38 µg Ag/ml [190-196]. Ahmad et al., developed silver nanoparticles using Sargentodoxa cuneata extract and these nanoparticles thus acted as reducing, capping and stabilizing agents and showed antileishmanial activity at IC50 values of 4.37 and 5.29 µg/ml, respectively [190-196]. In another research, Zahir et al., found out high antileishmanial efficacy of developed silver nanoparticles against Leishmania parasites at IC50 of 14.94 µg/ml and 3.89 µg/ml [190-196].

Gold Nanoparticles [GNPs]

Sazgarnia et al., reported the increased exposure time of the microwave to the parasites in the presence of GNPs induced a significant decline in promastigotes survival rate in comparison to similar samples without GNPs. The least survival of amastigotes was also recorded in the groups containing GNPs [204].

Helminth

Helminth also commonly known as parasitic worms which are invertebrate, elongated, round or flat bodies. Helminth infection causes morbidity and mortality. Helminth infections are generally treated using anthelmintics agents but some of these infections offer resistance towards drug. Metal-based nanoparticles like silver, gold and zinc oxide were used for the treatment of Helminth infections.

Rashid et al., reported that silver nanoparticles which were synthesized from momordica charantia plant extract and coated with polyaniline had a kill time of worms as 35.12±0.5 and 59.3±0.3 min [200-201].

Kar et al., reported that Gold nanoparticles prepared with the help of gold chloride and mycelia-free fungus affected the parasite and cause paralysis resulting in death [205].

Iron oxide nanoparticles were more effective than zinc oxide nanoparticles when treated against T. vitulorum and evaluated for the antihelmintic activity. Treatment with low dose of 0.004%, results in an increase of SOD.

Table 5: Various metal-based nanoparticles with anti-parasitic efficacy with therapeutic results

Metal nanoparticles Infection Therapeutic outcome References
Silver nanoparticles Malaria Inhibition of the growth of P. falciparum in vivo and in vitro [185-188]
Silver nanoparticles Malaria The AgNPs showed antiplasmodial activity against P. falciparum [189]
Silver nanoparticles Leishmaniasis Inhibition of proliferation and metabolic activity of promastigotes. Good antileishmanial activity in vitro and in vivo [190-196]
Silver nanoparticles Leishmaniasis Ag-NPs demonstrated significant antileishmanial effects by inhibiting the proliferation and metabolic activity of promastigotes. [197, 198]
Silver nanoparticles Leishmaniasis The IC50 for nanosilver solutions was high significantly [14.9 μg ml-1]. [199]
Silver nanoparticles Helminth Enhanced anthelmintic activity against worm [200, 201]
Gold nanoparticles Malaria Moderate delayed parasitemia rise in vivo, moderate antiplasmodial activity against P. falciparum [202, 203]
Gold nanoparticles Leishmaniasis The presence of GNPs during MW irradiation was more lethal for promastigotes and amastigotes in comparison to MW alone. [204]
Gold nanoparticles Helminth Affected the physiological functioning of the parasite causing paralysis and subsequent death [205]
Selenium nanoparticle Leishmaniasis Unlike selenium NPs, showed an anti-Leishmanial effect in vivo. [206]
Copper [II] nanoparticle Malaria The two compounds showed significant antimalarial activities against the parasites [207]
Zinc oxide nanoparticles and iron oxide nanoparticles Helminth The anthelmintic activity of the metal oxides nanoparticles was via induction of oxidative stress [208]

Future prospects

Despite the above-mentioned clinical efficacy of inorganic metal nanoparticles, it is extremely necessary to develop cost-effective metal nanoparticles so that application of these inorganic nanoparticles can increase further to treat infections than other drugs Also, there is a more need to study the toxicological properties and pharmacological properties for the treatment of all infection. Metal-based nanoparticles have the ability to overcome drug resistance which makes it promising for the treatment of infectious diseases in the future of medical sciences.

CONCLUSION

Infection diseases were classified as bacteria, fungi, viral and parasitic as well as their treatments is affected by drug resistance. Nowadays, most of the medicines used for the treatment of infections are suffering from drug toxicity, which are non-selective, and these infections can be overcome by metal-based nanoparticles.

They can also be engineered by introducing selected biological moieties with specific binding activity to selected target cells thereby improving their therapeutic efficacy at the pathological site. Metal-based nanoparticles of antimicrobial activities is related to their ability to produce ROS that damage the bacterial cell wall and bind to DNA or RNA, due to disturbed the microbial replication process, mitochondrial function as well as bacterial enzyme activity. The combinations of antibiotics with nanoparticles are exhibits the good synergistic effects against the microbes.

ACKNOWLEDGEMENT

I wish to express my sincere acknowledgment to Dr. Ashok Kumar Chauhan, President, RBEF parent organization of Amity University Madhya Pradesh [AUMP], Dr. Aseem Chauhan, Additional President, RBEF and chairman of Amity University Gwalior Campus, Lt. Gen. V. K. Sharma, AVSM [Retd.], Vice-Chancellor of AUMP Gwalior Campus and Head of Biotechnology Department, Invertis University for their valuable support and encouragement throughout the work.

FUNDING

Nil

AUTHOR CONTRIBUTION

MG wrote the manuscript draft and design the concept and finalized the manuscript.

CONFLICT OF INTERESTS

The authors confirm they have no conflict of interests.

REFERENCES

  1. Morens DM, Fauci AS. Emerging infectious diseases: threats to human health and global stability. PLOS Pathog 2013;9:e1003467.

  2. Fauci AS, Morens DM. The perpetual challenge of infectious diseases. N Eng J Med 2012;366:454–61.

  3. Parrish CR, Holmes EC, Morens DM, Park EC, Burke DS, Calisher CH, et al. Cross-species virus transmission and the emergence of new epidemic diseases. Microbiol Mol Biol Rev 2008;72:457–70.

  4. National Institutes of Health [US]. Biological sciences curriculum study NIH curriculum supplement series. In Understanding Emerging and Re-emerging Infectious Diseases; National Institutes of Health [US]: Bethesda, MD, USA; 2007. Available from: https://www.ncbi.nlm.nih.gov/books/NBK20370/ [Last accessed on 13 Mar 2018]

  5. Casadevall A. Antibody-based therapies for emerging infectious diseases. Emerg Infect Dis 1996;2:200.

  6. Saylor C, Dadachova E, Casadevall A. Monoclonal antibody-based therapies for microbial diseases. Vaccine 2009;27:G38–46.

  7. Mody VV, Siwale R, Singh A, Mody HR. Introduction to metallic nanoparticles. J Pharm Bioall Sci 2010;2:282–9.

  8. Pelgrift RY, Friedman AJ. Nanotechnology as a therapeutic tool to combat microbial resistance. Adv Drug Delivery Rev 2013;65:1803–15.

  9. Zhang L, Pornpattananangkul D, Hu CM J, Huang CM. Development of nanoparticles for antimicrobial drug delivery. Curr Med Chem 2010;6:585–94.

  10. Baek YW, An YJ. Microbial toxicity of metal oxide nanoparticles [CuO, NiO, ZnO, and Sb2O3] to Escherichia coli, Bacillus subtilis, and Streptococcus aureus. Sci Total Environ 2011;8:1603–8.

  11. Nath D, Banerjee P. Green nanotechnology-a new hope for medical biology. Environ Toxicol Pharmacol 2013;3:997–1014.

  12. Huh AJ, Kwon YJ. Nanoantibiotics: a new paradigm for treating infectious diseases using nanomaterials in the antibiotics resistant era. J Controlled Release 2011;2:128–45.

  13. Gatoo MA, Naseem S, Arfat MY, Mahmood Dar A, Qasim K, Zubair S. Physicochemical properties of nanomaterials: implication in associated toxic manifestations. BioMed Res Int 2014;1:8.

  14. Ashkarran AA, Ghavami M, Aghaverdi H, Stroeve P, Mahmoudi M. Bacterial effects and protein corona evaluations: crucial ignored factors in the prediction of bio-efficacy of various forms of silver nanoparticles. Chem Res Toxicol 2012;18:1231-42.

  15. Hajipour MJ, Fromm KM, Ashkarran AA, de Aberasturi DJ, de Larramendi IR, Rojo T, et al. Antibacterial properties of nanoparticles. Trends Biotechnol 2012;10:499-511.

  16. Blecher K, Nasir A, Friedman A. The growing role of nanotechnology in combating infectious disease. Virulence 2011;5:395-401.

  17. Vallyathan V, Shi X. The role of oxygen free radicals in occupational and environmental lung diseases. Environ Health Perspect 1997;1:165-77.

  18. Bonner JC. Lung fibrotic responses to particle exposure. Toxicol Pathol 2007;1:148-53.

  19. Ray PC, Yu H, Fu PP. Toxicity and environmental risks of nanomaterials: challenges and future needs. J Environ Sci Health Part C: Environ Carcinog Ecotoxicol Rev 2009;1:1-35.

  20. Nel A, Xia T, Mädler L, Li N. Toxic potential of materials at the nanolevel. Science 2006;5761:622-7.

  21. Xia T, Kovochich M, Liong M, Madler L, Gilbert B, Shi H, et al. Comparison of the mechanism of toxicity of zinc oxide and cerium oxide nanoparticles based on dissolution and oxidative stress properties. ACS Nano 2008;10:2121-34.

  22. Wang S, Lu W, Tovmachenko O, Rai US, Yu H, Ray PC. Challenge in understanding size and shape-dependent toxicity of gold nanomaterials in human skin keratinocytes. Chem Phys Lett 2008;1-3:145-9.

  23. Shaligram S, Campbell A. Toxicity of copper salts is dependent on solubility profile and cell type tested. Toxicol In Vitro 2013;2:844-51.

  24. Lu W, Senapati D, Wang S, Tovmachenko O, Singh AK, Yu H, et al. Effect of surface coating on the toxicity of silver nanomaterials on human skin keratinocytes. Chem Phys Lett 2010;1-3:92-6.

  25. Huh AJ, Kwon YJ. “Nanoantibiotics”: a new paradigm for treating infectious diseases using nanomaterials in the antibiotics resistant era. J Controlled Release 2011;2:128-45.

  26. Pelgrift RY, Friedman AJ. Nanotechnology as a therapeutic tool to combat microbial resistance. Adv Drug Delivery Rev 2013;13-14:1803-15.

  27. Nathan C, Cunningham Bussel A. Beyond oxidative stress: an immunologist's guide to reactive oxygen species. Nat Rev Immunol 2013;5:349-61.

  28. Pan X, Redding JE, Wiley PA, Wen L, McConnell JS, Zhang B. Mutagenicity evaluation of metal oxide nanoparticles by the bacterial reverse mutation assay. Chemosphere 2010;1:113-6.

  29. Wang S, Lawson R, Ray PC, Yu H. Toxic effects of gold nanoparticles on salmonella typhimurium bacteria. Toxicol Ind Health 2011;6:547-54.

  30. Matejka V, Tokarsky J. Photocatalytical nanocomposites: a review. J Nanosci Nanotechnol 2014;2:1597-616.

  31. Maness PC, Smolinski S, Blake DM, Huang Z, Wolfrum EJ, Jacoby WA. Bactericidal activity of photocatalytic TiO2 reaction: toward an understanding of its killing mechanism. Appl Environ Microbiol 1999;9:4094-8.

  32. Beyth N, Yudovin Farber I, Perez Davidi M, Domb AJ, Weiss EI. Polyethyleneimine nanoparticles incorporated into resin composite cause cell death and trigger biofilm stress in vivo. Proc Natl Acad Sci India 2010;51:22038-43.

  33. Lok CN, Ho CM, Chen R, He QY, Yu WY, Sun H, et al. Silver nanoparticles: partial oxidation and antibacterial activities. J Biol Inorg Chem 2007;4:527-34.

  34. Mohamed MM, Fouad SA, Elshoky HA, Mohammed GM, Salaheldin TA. Antibacterial effect of gold nanoparticles against corynebacterium pseudotuberculosis. Int J Vet Sci Med 2017;1:23-9.

  35. Gong P, Li H, He X, Wang K, Hu J, Tan W, et al. Preparation and antibacterial activity of Fe3O4@ Ag nanoparticles. Nanot 2007;28:285-604.

  36. Stoimenov PK, Klinger RL, Marchin GL, Klabunde KJ. Metal oxide nanoparticles as bactericidal agents. Langmuir 2002;17:6679-86.

  37. Xie Y, He Y, Irwin PL, Jin T, Shi X. Antibacterial activity and mechanism of action of zinc oxide nanoparticles against Campylobacter jejuni. Appl Environ Microbiol 2011;7:2325-31.

  38. Yoon KY, Byeon JH, Park JH, Hwang J. Susceptibility constants of Escherichia coli and Bacillus subtilis to silver and copper nanoparticles. Sci Total Environ 2007;2-3:572-5.

  39. Bonnet M, Massard C, Veisseire P, Camares O, Awitor KO. Environmental toxicity and antimicrobial efficiency of titanium dioxide nanoparticles in suspension. J Biomater Nanobiotechnol 2015;3:11.

  40. Simon Deckers A, Loo S, Mayne L’hermite M, Herlin Boime N, Menguy N, Reynaud C, et al. Size-, composition-and shape-dependent toxicological impact of metal oxide nanoparticles and carbon nanotubes toward bacteria. Environ Sci Technol 2009;21:8423-9.

  41. Tallury P, Malhotra A, Byrne LM, Santra S. Nanobioimaging and sensing of infectious diseases. Adv Drug Delivery Rev 2010;4-5:424-37.

  42. Baldoni D. Innovative methods for the diagnosis and treatment of implant-associated infections. Doctoral dissertation, University_of_Basel; 2009.

  43. Ribet D, Cossart P. How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes Infect 2015;3:173-83.

  44. Melville S, Craig L. Type IV pili in gram-positive bacteria. Microbiol Mol Biol Rev 2013;3:323-41.

  45. Pizarro Cerda J, Cossart P. Bacterial adhesion and entry into host cells. Cell 2006;4:715-27.

  46. Hall Stoodley L, Costerton JW, Stoodley P. Bacterial biofilms: from the natural environment to infectious diseases. Nat Rev Microbiol 2004;2:95-108.

  47. Joo HS, Otto M. Molecular basis of in vivo biofilm formation by bacterial pathogens. Chem Biol 2012;12:1503-13.

  48. Jafari AR, Mosavi T, Mosavari N, Majid A, Movahedzade F, Tebyaniyan M, et al. Mixed metal oxide nanoparticles inhibit growth of Mycobacterium tuberculosis into THP-1 cells. Int J Mycobact 2016;5:S181-3.

  49. Praba VL, Kathirvel M, Vallayyachari K, Surendar K, Muthuraj M, Jesuraj PJ, et al. Bactericidal effect of silver nanoparticles against mycobacterium tuberculosis. J Bionanosci 2013;3:282-7.

  50. Singh R, Nawale LU, Arkile M, Shedbalkar UU, Wadhwani SA, Sarkar D, et al. Chemical and biological metal nanoparticles as antimycobacterial agents: a comparative study. Int J Antimicrob Agents 2015;2:183-8.

  51. Inbaneson SJ, Ravikumar S, Manikandan N. Antibacterial potential of silver nanoparticles against isolated urinary tract infectious bacterial pathogens. Appl Nanosci 2011;4:231-6.

  52. Pickard R, Lam T, MacLennan G, Starr K, Kilonzo M, McPherson G, et al. Antimicrobial catheters for reduction of symptomatic urinary tract infection in adults requiring short-term catheterization in hospital: a multicentre randomized controlled trial. Lancet 2012;9857:1927-35.

  53. Ritter J, Thomas L, Lederer J, Jarvis WR. Effectiveness of a silver-alloy and hydrogel coated urinary catheter on symptomatic catheter-associated urinary tract infections. Am J Infect Control 2013;6:S143-44.

  54. Ford J, Hughes G, Phillips P. Literature review of silver-coated urinary catheters-draft SMTL; 2014. Available from: http://www.medidex.com/research/830-silver-coated-catheters-full-article.html [Last accessed on 07 Jul 2017]

  55. Thibon P, Le Coutour X, Leroyer R, Fabry J. Randomized multi-center trial of the effects of a catheter coated with hydrogel and silver salts on the incidence of hospital-acquired urinary tract infections. J Hosp Infect 2000;2:117-24.

  56. Srinivasan A, Karchmer T, Richards A, Song X, Perl TM. A prospective trial of a novel, silicone-based, silver-coated foley catheter for the prevention of nosocomial urinary tract infections. Infect Controlled Hosp Epidemiol 2006;1:38-43.

  57. Akram FE, El-Tayeb T, Abou-Aisha K, El-Azizi M. A combination of silver nanoparticles and visible blue light enhances the antibacterial efficacy of ineffective antibiotics against methicillin-resistant staphylococcus aureus [MRSA]. Ann Clin Microbiol Antimicrob 2016;1:48.

  58. Fatimah I. Green synthesis of silver nanoparticles using extract of Parkia speciosa Hassk pods assisted by microwave irradiation. J Adv Res 2016;6:961-9.

  59. Verma A, Mehata MS. Controllable synthesis of silver nanoparticles using neem leaves and their antimicrobial activity. J Radiat Res Appl Sci 2016;1:109-15.

  60. Tripathy A, Raichur AM, Chandrasekaran N, Prathna TC, Mukherjee A. Process variables in the biomimetic synthesis of silver nanoparticles by aqueous extract of Azadirachta Indica [Neem] leaves. J Nanopar Res 2010;1:237-46.

  61. Krishnaraj C, Jagan EG, Rajasekar S, Selvakumar P, Kalaichelvan PT, Mohan NJ. Synthesis of silver nanoparticles using acalypha indica leaf extracts and its antibacterial activity against water-borne pathogens. Colloids Surf B 2010;1:50-6.

  62. Ponarulselvam S, Panneerselvam C, Murugan K, Aarthi N, Kalimuthu K, Thangamani S. Synthesis of silver nanoparticles using leaves of catharanthus roseus linn. G. don and their antiplasmodial activities. Asian Pac J Trop Biomed 2012;7:574.

  63. Tippayawat P, Phromviyo N, Boueroy P, Chompoosor A. Green synthesis of silver nanoparticles in aloe vera plant extract prepared by a hydrothermal method and their synergistic antibacterial activity. Peer J 2016;4:e2589.

  64. Ali ZA, Yahya R, Sekaran SD, Puteh R. Green synthesis of silver nanoparticles using apple extract and its antibacterial properties. Adv Mater Sci Eng 2016;1. https://doi.org/10.1155/2016/4102196

  65. Allafchian AR, Mirahmadi Zare SZ, Jalali SA, Hashemi SS, Vahabi MR. Green synthesis of silver nanoparticles using phlomis leaf extract and investigation of their antibacterial activity. J Nanostruct Chem 2016;2:129-35.

  66. Maiti S, Krishnan D, Barman G, Ghosh SK, Laha JK. Antimicrobial activities of silver nanoparticles synthesized from Lycopersicon esculentum extract. J Anal Sci Technol 2014;1:1-7.

  67. Chiguvare H, Oyedeji OO, Matewu R, Aremu O, Oyemitan IA, Oyedeji AO, et al. Synthesis of silver nanoparticles using Buchu plant extracts and their analgesic properties. Molecules 2016;6:774.

  68. Singhal G, Bhavesh R, Kasariya K, Sharma AR, Singh RP. Biosynthesis of silver nanoparticles using ocimum sanctum [Tulsi] leaf extract and screening its antimicrobial activity. J Nanopart Res 2011;7:2981-8.

  69. Nabikhan A, Kandasamy K, Raj A, Alikunhi NM. Synthesis of antimicrobial silver nanoparticles by callus and leaf extracts from saltmarsh plant, sesuvium portulacastrum L. Colloids Surf B 2010;2:488-93.

  70. Das VL, Thomas R, Varghese RT, Soniya EV, Mathew J, Radhakrishnan EK. Extracellular synthesis of silver nanoparticles by the bacillus strain CS 11 isolated from the industrialized area. 3 Biotech 2014;2:121-6.

  71. Choi O, Deng KK, Kim NJ, Ross Jr L, Surampalli RY, Hu Z. The inhibitory effects of silver nanoparticles, silver ions, and silver chloride colloids on microbial growth. Water Res 2008;12:3066-74.

  72. Feng QL, Wu J, Chen GQ, Cui FZ, Kim TN, Kim JO. A mechanistic study of the antibacterial effect of silver ions on escherichia coli and staphylococcus aureus. J Biomed Mat Res 2000;4:662-8.

  73. Sondi I, Salopek Sondi B. Silver nanoparticles as antimicrobial agent: a case study on E. coli as a model for gram-negative bacteria. J Colloid Interface Sci 2004;1:177-82.

  74. Prabhu S, Poulose EK. Silver nanoparticles: mechanism of antimicrobial action, synthesis, medical applications, and toxicity effects. Int Nano Lett 2012;1:32.

  75. Kim JS, Kuk E, Yu KN, Kim JH, Park SJ, Lee HJ, et al. Antimicrobial effects of silver nanoparticles. Nanomedicine 2007;1:95-101.

  76. Lu Z, Rong K, Li J, Yang H, Chen R. Size-dependent antibacterial activities of silver nanoparticles against oral anaerobic pathogenic bacteria. J Mater Sci: Mater Med 2013;6:1465-71.

  77. Morones JR, Elechiguerra JL, Camacho A, Holt K, Kouri JB, Ramirez JT, et al. The bactericidal effect of silver nanoparticles. Nanotech 2005;10:2346.

  78. Singh T, Jyoti K, Patnaik A, Singh A, Chauhan R, Chandel SS. Biosynthesis, characterization and antibacterial activity of silver nanoparticles using an endophytic fungal supernatant of raphanus sativus. J Genet Eng Biotechnol 2017;15:31-9.

  79. El-Sheekh MM, El-Kassas HY. Algal production of nano-silver and gold: Their antimicrobial and cytotoxic activities: a review. J Genet Eng Biotechnol 2016;2:299-310.

  80. Saravanan C, Rajesh R, Kaviarasan T, Muthukumar K, Kavitake D, Shetty PH. Synthesis of silver nanoparticles using bacterial exopolysaccharide and its application for degradation of azo-dyes. Biotechnol Rep 2017;5:33-40.

  81. Narayanan KB, Sakthivel N. Biological synthesis of metal nanoparticles by microbes. Adv Colloid Interface Sci 2010;1-2:1-3.

  82. Ashajyothi C, Oli AK, Prabhurajeshwar C. Potential bactericidal effect of silver nanoparticles synthesized from enterococcus species. Orient J Chem 2014;3:1253-62.

  83. Minaeian S, Shahverdi AR, Nouhi AA, Shahverdi HR. Extracellular biosynthesis of silver nanoparticles by some bacteria. J Sci IAU 2008;17:1–4.

  84. Shirley AD, Dayanand A, Sreedhar B, Dastager SG. Antimicrobial activity of silver nanoparticles synthesized from novel streptomyces species. Dig J Nanomater Bios 2010;2:447-51.

  85. Wan G, Ruan L, Yin Y, Yang T, Ge M, Cheng X. Effects of silver nanoparticles in combination with antibiotics on the resistant bacteria acinetobacter baumannii. Int J Nanomed 2016;11:3789.

  86. Li P, Li J, Wu C, Wu Q, Li J. Synergistic antibacterial effects of β-lactam antibiotic combined with silver nanoparticles. Nanotech 2005;9:1912.

  87. Le AT, Le TT, Tran HH, Dang DA, Tran QH, Vu DL. Powerful colloidal silver nanoparticles for the prevention of gastrointestinal bacterial infections. Adv Nat Sci: Nanosci Nanotechnol 2012;4:045007.

  88. Sadiq IM, Chowdhury B, Chandrasekaran N, Mukherjee A. Antimicrobial sensitivity of Escherichia coli to alumina nanoparticles. Nanomedicine 2009;3:282-6.

  89. Jalal M, Ansari MA, Shukla AK, Ali SG, Khan HM, Pal R, et al. Green synthesis and antifungal activity of Al 2 O 3 NPs against fluconazole-resistant Candida spp isolated from a tertiary care hospital. RSC Adv 2016;109:107577-90.

  90. Ansari MA, Khan HM, Alzohairy MA, Jalal M, Ali SG, Pal R, et al. Green synthesis of Al 2 O 3 nanoparticles and their bactericidal potential against clinical isolates of multi-drug resistant Pseudomonas aeruginosa. World J Microbiol Biotechnol 2015;1:153-64.

  91. Prashanth PA, Raveendra RS, Hari Krishna R, Ananda S, Bhagya NP, Nagabhushana BM, et al. Synthesis, characterizations, antibacterial and photoluminescence studies of solution combustion-derived α-Al2O3 nanoparticles. J Asian Ceram Soc 2015;3:345-51.

  92. Brintha SR, Ajitha M. Synthesis, structural and antibacterial activity of aluminium and nickel doped ZnO nanoparticles by sol-gel method. Asian J Chem Sci 2016;12:1-9.

  93. Ahmed KB, Subramanian S, Sivasubramanian A, Veerappan G, Veerappan A. Preparation of gold nanoparticles using salicornia brachiata plant extract and evaluation of the catalytic and antibacterial activity. Spectrochim Acta Part A 2014;130:54-8.

  94. Zhang Y, Shareena Dasari TP, Deng H, Yu H. Antimicrobial activity of gold nanoparticles and ionic gold. J Environ Sci Health Part C Environ Carcinog Ecotoxicol Rev 2015;3:286-327.

  95. Rai A, Prabhune A, Perry CC. Antibiotic mediated synthesis of gold nanoparticles with potent antimicrobial activity and their application in antimicrobial coatings. J Mater Chem 2010;32:6789-90.

  96. Saha B, Bhattacharya J, Mukherjee A, Ghosh A, Santra C, Dasgupta AK, et al. In vitro structural and functional evaluation of gold nanoparticles conjugated antibiotics. Nanoscale Res Lett 2007;12:614.

  97. Bagga P, Ansari TM, Siddiqui HH, Syed A, Bahkali AH, Rahman MA, et al. Bromelain capped gold nanoparticles as the novel drug delivery carriers to aggrandize the effect of the antibiotic levofloxacin. EXCLI J 2016;15:772.

  98. Li X, Robinson SM, Gupta A, Saha K, Jiang Z, Moyano DF, et al. Functional gold nanoparticles as potent antimicrobial agents against multi-drug-resistant bacteria. ACS Nano 2014;10:10682-6.

  99. Adavallan K, Krishnakumar N. Mulberry leaf extract mediated synthesis of gold nanoparticles and its antibacterial activity against human pathogens. Adv Nat Sci: Nanosci Nanotechnol 2014;2:025018.

  100. Taran M, Rad M, Alavi M. Antibacterial activity of copper oxide [CuO] nanoparticles biosynthesized by bacillus sp. FU4: optimization of experiment design. Pharm Sci 2017;3:198-206.

  101. Sutradhar P, Saha M, Maiti D. Microwave synthesis of copper oxide nanoparticles using tea leaf and coffee powder extracts and its antibacterial activity. J Nanostruct Chem 2014;1:86.

  102. Altikatoglu M, Attar A, Erci F, Cristache CM, Isildak I. Green synthesis of copper oxide nanoparticles using ocimum basilicum extract and their antibacterial activity. Fresenius Environ Bull 2017;12:7832-7.

  103. Azam A, Ahmed AS, Oves M, Khan MS, Memic A. Size-dependent antimicrobial properties of CuO nanoparticles against gram-positive and-negative bacterial strains. Int J Nanomed 2012;7:3527.

  104. Chatterjee AK, Chakraborty R, Basu T. Mechanism of antibacterial activity of copper nanoparticles. Nanotech 2014;13:135101.

  105. Kumar PV, Shameem U, Kollu P, Kalyani RL, Pammi SV. Green synthesis of copper oxide nanoparticles using Aloe vera leaf extract and its antibacterial activity against fish bacterial pathogens. BioNanoSci 2015;3:135-9.

  106. Devatha CP, Jagadeesh K, Patil M. Effect of green synthesized iron nanoparticles by Azardirachta Indica in different proportions on antibacterial activity. Environ Nanotechnol Monit Manag 2018;9:85-94.

  107. Kanagasubbulakshmi S, Kadirvelu K. Green synthesis of iron oxide nanoparticles using lagenaria siceraria and evaluation of its antimicrobial activity. Def Life Sci J 2017;4:422–7.

  108. Rafi MM, Ahmed KS, Nazeer KP, Kumar DS, Thamilselvan M. Synthesis, characterization and magnetic properties of hematite [α-Fe 2 O 3] nanoparticles on polysaccharide templates and their antibacterial activity. Appl Nanosci 2015;4:515-20.

  109. Masadeh MM, Karasneh GA, Al-Akhras MA, Albiss BA, Aljarah KM, Al-Azzam SI, et al. Cerium oxide and iron oxide nanoparticles abolish the antibacterial activity of ciprofloxacin against gram-positive and gram-negative biofilm bacteria. Cytotechnology 2015;3:427-35.

  110. Narayanasamy P, Switzer BL, Britigan BE. Prolonged-acting, multi-targeting gallium nanoparticles potently inhibit the growth of both HIV and mycobacteria in co-infected human macrophages. Sci Rep 2015;1:1-7.

  111. Kurtjak M, Vukomanovic M, Kramer L, Suvorov D. Biocompatible nano-gallium/hydroxyapatite nanocomposite with antimicrobial activity. J Mater Sci Mater Med 2016;11:170.

  112. Choi SR, Britigan BE, Moran DM, Narayanasamy P. Gallium nanoparticles facilitate phagosome maturation and inhibit growth of virulent mycobacterium tuberculosis in macrophages. PLoS One 2017;5:e0177987.

  113. Olakanmi O, Gunn JS, Su S, Soni S, Hassett DJ, Britigan BE. Gallium disrupts iron uptake by intracellular and extracellular Francisella strains and exhibits therapeutic efficacy in a murine pulmonary infection model. Antimicrob Agents Chemother 2010;1:244-53.

  114. Sharma G, Soni R, Jasuja ND. Phytoassisted synthesis of magnesium oxide nanoparticles with swertia chirayaita. J Taibah Univ Sci 2017;11:471-7.

  115. Besinis A, De Peralta T, Handy RD. The antibacterial effects of silver, titanium dioxide and silica dioxide nanoparticles compared to the dental disinfectant chlorhexidine on streptococcus mutans using a suite of bioassays. Nanotoxicology 2014;1:1-6.

  116. Alhadrami HA, Al-Hazmi F. Antibacterial activities of titanium oxide nanoparticles. J Bioelectron Nanotechnol 2017;2:5.

  117. Desai VS, Meenal K. Antimicrobial activity of titanium dioxide nanoparticles synthesized by sol-gel technique. Res J Microbiol 2009;3:97-103.

  118. Lin X, Li J, Ma S, Liu G, Yang K, Tong M, et al. Toxicity of TiO 2 nanoparticles to Escherichia coli: effects of particle size, crystal phase and water chemistry. PloS One 2014;10:e110247.

  119. Planchon M, Ferrari R, Guyot F, Gelabert A, Menguy N, Chaneac C, et al. Interaction between Escherichia coli and TiO2 nanoparticles in natural and artificial waters. Colloids Surf B 2013;102:158-64.

  120. Tong T, Binh CT, Kelly JJ, Gaillard JF, Gray KA. Cytotoxicity of commercial nano-TiO2 to Escherichia coli assessed by high-throughput screening: effects of environmental factors. Water Res 2013;7:2352-62.

  121. Jesline A, John NP, Narayanan PM, Vani C, Murugan S. Antimicrobial activity of zinc and titanium dioxide nanoparticles against biofilm-producing methicillin-resistant Staphylococcus aureus. Appl Nanosci 2015;2:157-62.

  122. Voicu G, Oprea O, Vasile BS, Andronescu E. Antibacterial activity of zinc oxide-gentamicin hybrid material. Dig J Nanomater Bios 2013;8:3.

  123. Bhuyan T, Mishra K, Khanuja M, Prasad R, Varma A. Biosynthesis of zinc oxide nanoparticles from Azadirachta indica for antibacterial and photocatalytic applications. Mater Sci Semicond Process 2015;32:55-61.

  124. Raghupathi KR, Koodali RT, Manna AC. Size-dependent bacterial growth inhibition and mechanism of antibacterial activity of zinc oxide nanoparticles. Langmuir 2011;7:4020-8.

  125. Hsueh YH, Ke WJ, Hsieh CT, Lin KS, Tzou DY, Chiang CL. ZnO nanoparticles affect bacillus subtilis cell growth and biofilm formation. PloS One 2015;6:e0128457.

  126. Fernandes S, Simoes M, Dias N, Santos C, Lima N. Fungicidal activity of microbicides. Russell, hugo and ayliffe’s: principles and practice of disinfection, preservation and sterilization. Wiley 2013;10:142-54.

  127. Pereira L, Dias N, Santos C, Lima N. The use of MALDI-TOF ICMS as an alternative tool for Trichophyton rubrum identification and typing. Enfermedades Infecciosasy Microbiol Clin 2014;1:11-7.

  128. Havlickova B, Czaika VA, Friedrich M. Epidemiological trends in skin mycoses worldwide. Mycoses 2008;51:2-15.

  129. Dias N, Santos C, Portela M, Lima N. Toenail onychomycosis in a portuguese geriatric population. Mycopathologia 2011:1:55-61.

  130. Freytes DM, Arroyo Novoa CM, Figueroa Ramos MI, Ruiz Lebron RB, Stotts NA, Busquets A. Skin disease in HIV-positive persons living in puerto rico. Adv Skin Wound Care 2007;3:149-56.

  131. Mukherjee PK, Leidich SD, Isham N, Leitner I, Ryder NS, Ghannoum MA. Clinical trichophyton rubrum strain exhibiting primary resistance to terbinafine. Antimicrob Agents Chemother 2003;1:82-6.

  132. Kim JS, Kuk E, Yu KN, Kim JH, Park SJ, Lee HJ, et al. Antimicrobial effects of silver nanoparticles. Nanomedicine 2007;1:95-101.

  133. Auberger J, Lass Florl C, Aigner M, Clausen J, Gastl G, Nachbaur D. Invasive fungal breakthrough infections, fungal colonization and emergence of resistant strains in high-risk patients receiving antifungal prophylaxis with posaconazole: real-life data from a single-centre institutional retrospective observational study. J Antimicrob Chem 2012;9:2268-73.

  134. Rai M, Gade A, Yadav A. Biogenic nanoparticles: an introduction to what they are, how they are synthesized and their applications. Metal Nanopart Microbiol Springer Berlin Heidelberg; 2011. p. 1-14.

  135. Mohanta YK, Panda SK, Jayabalan R, Sharma N, Bastia AK, Mohanta TK. Antimicrobial, antioxidant and cytotoxic activity of silver nanoparticles synthesized by leaf extract of Erythrina suberosa [Roxb.]. Front Mol Biosci 2017;17:14.

  136. Lara HH, Romero Urbina DG, Pierce C, Lopez Ribot JL, Arellano Jimenez MJ, Jose Yacaman M. Effect of silver nanoparticles on candida albicans biofilms: an ultrastructural study. J Nanobiotechnol 2015;1:1-2.

  137. Chwalibog A, Sawosz E, Hotowy A, Szeliga J, Mitura S, Mitura K, et al. Visualization of interaction between inorganic nanoparticles and bacteria or fungi. Int J Nanomed 2010;5:1085.

  138. Monteiro DR, Gorup LF, Silva S, Negri M, De Camargo ER, Oliveira R, et al. Silver colloidal nanoparticles: antifungal effect against adhered cells and biofilms of candida albicans and candida glabrata. Biofouling 2011;7:711-9.

  139. Nasrollahi A, Pourshamsian KH, Mansourkiaee P. Antifungal activity of silver nanoparticles on some of fungi. Int J Nano Dim 2011;1:233–9.

  140. Hwang IS, Lee J, Hwang JH, Kim KJ, Lee DG. Silver nanoparticles induce apoptotic cell death in candida albicans through the increase of hydroxyl radicals. FEBS J 2012;7:1327-38.

  141. Rajarathinam M, Kalaichelvan PT. Biogenic nanosilver as a potential antibacterial and antifungal additive to commercially available dish wash and hand wash for an enhanced antibacterial and antifungal activity against selected pathogenic strains. Int Res J Pharma 2013;4:68-75.

  142. Ahmad A, Wei Y, Syed F, Tahir K, Taj R, Khan AU, et al. Amphotericin B-conjugated biogenic silver nanoparticles as an innovative strategy for fungal infections. Microb Pathog 2016;99:271-81.

  143. Falkiewicz Dulik M, Macura AB. Nanosilver as substance biostabilising footwear materials in the foot mycosis prophylaxis. Mikologia Lekarska 2008;15:145-50.

  144. Noorbakhsh F, Rezaie S, Shahverdi AR. Antifungal effects of silver nanoparticle alone and with combination of antifungal drug on dermatophyte pathogen Trichophyton rubrum. Int Conf Biosci Biochem Bioinfo 2011;5:364-7.

  145. Marcato PD, Duran M, Huber SC, Rai M, Melo PS, Alves OL, Duran N. Biogenic silver nanoparticles and its antifungal activity as a new topical transungual drug. J Nano Res 2012;20:99-107.

  146. Weitz IS, Maoz M, Panitz D, Eichler S, Segal E. Combination of CuO nanoparticles and fluconazole: preparation, characterization, and antifungal activity against candida albicans. J Nanopart Res 2015;8:342.

  147. Sojinrin T, Conde J, Liu K, Curtin J, Byrne HJ, Cui D, et al. Plasmonic gold nanoparticles for detection of fungi and human cutaneous fungal infections. Anal Bioanal Chem 2017;19:4647-58.

  148. Niemirowicz K, Durnas B, Tokajuk G, Piktel E, Michalak G, Gu X, et al. Formulation and candidacidal activity of magnetic nanoparticles coated with cathelicidin LL-37 and ceragenin CSA-13. Sci Rep 2017;1:1-2.

  149. Seddighi NS, Salari S, Izadi AR. Evaluation of antifungal effect of iron-oxide nanoparticles against different candida species. IET Nanobiotechnol 2017;7:883-8.

  150. Haghighi F, Roudbar Mohammadi S, Mohammadi P, Hosseinkhani S, Shipour R. Antifungal activity of TiO2 nanoparticles and EDTA on Candida albicans biofilms. Infect Epidemiol Microbiol 2013;1:33-8.

  151. Sardella D, Gatt R, Valdramidis VP. Assessing the efficacy of zinc oxide nanoparticles against penicillium expansum by automated turbidimetric analysis. Myco 2018;1:43-8.

  152. He L, Liu Y, Mustapha A, Lin M. Antifungal activity of zinc oxide nanoparticles against Botrytis cinerea and penicillium expansum. Microbiol Res 2011;3:207-15.

  153. Xue J, Luo Z, Li P, Ding Y, Cui Y, Wu Q. A residue-free green synergistic antifungal nanotechnology for pesticide thiram by ZnO nanoparticles. Sci Rep 2014;4:5408.

  154. Shinde SS. Antimicrobial activity of ZnO nanoparticles against pathogenic bacteria and fungi. Sci Med Central 2015;3:1033.

  155. Auyeung A, Casillas Santana MA, Martinez Castanon GA, Slavin YN, Zhao W, Asnis J, et al. Effective control of molds using a combination of nanoparticles. PloS One 2017;1:e0169940.

  156. Arciniegas Grijalba PA, Patino Portela MC, Mosquera Sanchez LP, Guerrero Vargas JA, Rodriguez Paez JE. ZnO nanoparticles [ZnO-NPs] and their antifungal activity against coffee fungus erythricium salmonicolor. Appl Nanosci 2017;5:225-41.

  157. Strasfeld L, Chou S. Antiviral drug resistance: mechanisms and clinical implications. Infect Dis Clin 2010;3:809-33.

  158. Global AIDS/HIV Epidermic. Available from: https://www.aids.gov/hiv-aids-basics/hiv-aids-101/global-statistics/ [Last accessed on 10 Mar 2018].

  159. Lara HH, Ayala Nunez NV, Ixtepan Turrent L, Rodriguez Padilla C. Mode of antiviral action of silver nanoparticles against HIV-1. J Nanobiotechnol 2010;1:1-10.

  160. Sun RW, Chen R, Chung NP, Ho CM, Lin CL, Che CM. Silver nanoparticles fabricated in Hepes buffer exhibit cytoprotective activities toward HIV-1 infected cells. Chem Comm 2005;40:5059-61.

  161. Elechiguerra JL, Burt JL, Morones JR, Camacho Bragado A, Gao X, Lara HH, et al. Interaction of silver nanoparticles with HIV-1. J Nanobiotechnol 2005;1:1-10.

  162. Baram Pinto D, Shukla S, Perkas N, Gedanken A, Sarid R. Inhibition of herpes simplex virus type 1 infection by silver nanoparticles capped with mercaptoethane sulfonate. Bioconjugate Chem 2009;8:1497-502.

  163. Hu RL, Li SR, Kong FJ, Hou RJ, Guan XL, Guo F. Inhibition effect of silver nanoparticles on herpes simplex virus 2. Genet Mol Res 2014;3:7022-8.

  164. Samji NS, Anand BS. Viral hepatitis; 2017. Available from: http://emedicine.medscape.com/article/775507-overview#a7 [Last accessed on 10 Mar 2018]

  165. Lu L, Sun RW, Chen R, Hui CK, Ho CM, Luk JM, et al. Silver nanoparticles inhibit hepatitis B virus replication. Antiviral Ther 2008;2:253.

  166. Davis CP. Available from: https://www.medicinenet.com/ viral_hepatitis/article.htm [Last accessed on 10 Mar 2018]

  167. Xiang D, Zheng Y, Duan W, Li X, Yin J, Shigdar S, et al. Inhibition of A/Human/Hubei/3/2005 [H3N2] influenza virus infection by silver nanoparticles in vitro and in vivo. Int J Nanomed 2013;8:4103.

  168. Mori Y, Ono T, Miyahira Y, Nguyen VQ, Matsui T, Ishihara M. Antiviral activity of silver nanoparticle/chitosan composites against H1N1 influenza a virus.
    Nanoscale Res Lett 2013;1:93.

  169. Li Y, Lin Z, Zhao M, Guo M, Xu T, Wang C, et al. Reversal of H1N1 influenza virus-induced apoptosis by silver nanoparticles functionalized with amantadine. RSC Adv 2016;92:89679-86.

  170. Sametband M, Shukla S, Meningher T, Hirsh S, Mendelson E, Sarid R, et al. Effective multi-strain inhibition of influenza virus by anionic gold nanoparticles. Med Chem Comm 2011;5:421-3.

  171. Xiang DX, Chen Q, Pang L, Zheng CL. Inhibitory effects of silver nanoparticles on H1N1 influenza a virus in vitro. J Virol Methods 2011;1-2:137-42.

  172. Mehrbod P, Motamed N, Tabatabaeian M, Soleymani ER, Amini E, Shahidi M, et al. In vitro antiviral effect of “Nanosilver” on influenza virus. DARU J Pharm Sci 2015;17:88–93.

  173. Kesarkar R, Oza G, Pandey S, Dahake R, Mukherjee S, Chowdhary A, et al. Gold nanoparticles: effective as both entry inhibitors and virus neutralizing agents against HIV. J Microbiol Biotechnol Res 2017;2:276–83.

  174. Sarid R, Gedanken A, Baram Pinto D. Inventors; Bar Ilan University, assignee. Pharmaceutical compositions comprising water-soluble sulfonate-protected nanoparticles and uses there of. United States patent US 8,758,777; 2014.

  175. Baram Pinto D, Shukla S, Gedanken A, Sarid R. Inhibition of HSV‐1 attachment, entry, and cell‐to‐cell spread by functionalized multivalent gold nanoparticles. Small 2010;9:1044-50.

  176. Yasri S, Wiwanitkit V. Effect of gold nanoparticle on viral load of hepatitis C virus. J Coast Life Med 2014;2:2754-61.

  177. Lee MY, Yang JA, Jung HS, Beack S, Choi JE, Hur W, et al. Hyaluronic acid–gold nanoparticle/interferon α complex for targeted treatment of hepatitis C virus infection. ACS Nano 2012;11:9522-31.

  178. Taubenberger JK, Morens DM. The pathology of influenza virus infections. Annu Rev Pathol Mech Dis 2008;3:499-522.

  179. Feng F, Sakoda Y, Ohyanagi T, Nagahori N, Shibuya H, Okamastu M, et al. Novel thiosialosides tethered to metal nanoparticles as potent influenza a virus haemagglutinin blockers. Antiviral Chem Chemother 2013;2:59-65.

  180. Ryoo SR, Jang H, Kim KS, Lee B, Kim KB, Kim YK, Yeo WS, et al. Functional delivery of DNAzyme with iron oxide nanoparticles for hepatitis C virus gene knockdown. Biomaterials 2012;9:2754-61.

  181. Hang X, Peng H, Song H, Qi Z, Miao X, Xu W. Antiviral activity of cuprous oxide nanoparticles against hepatitis C virus in vitro. J Virol Methods 2015;222:150-7.

  182. Antoine TE, Hadigal SR, Yakoub AM, Mishra YK, Bhattacharya P, Haddad C, et al. Intravaginal zinc oxide tetrapod nanoparticles as novel immunoprotective agents against genital herpes. J Immunol 2016;11:4566-75.

  183. Mishra YK, Adelung R, Rohl C, Shukla D, Spors F, Tiwari V. Virostatic potential of micro–nano filopodia-like ZnO structures against herpes simplex virus-1. Antiviral Res 2011;2:305-12.

  184. Mishra A, Kaushik NK, Sardar M, Sahal D. Evaluation of antiplasmodial activity of green synthesized silver nanoparticles. Colloids Surf B 2013;111:713-8.

  185. Jaganathan A, Murugan K, Panneerselvam C, Madhiyazhagan P, Dinesh D, Vadivalagan C, et al. Earthworm-mediated synthesis of silver nanoparticles: a potent tool against hepatocellular carcinoma, Plasmodium falciparum parasites and malaria mosquitoes. Parasitol Int 2016;3:276-84.

  186. Murugan K, Panneerselvam C, Subramaniam J, Madhiyazhagan P, Hwang JS, Wang L, et al. Eco-friendly drugs from the marine environment: spongeweed-synthesized silver nanoparticles are highly effective on plasmodium falciparum and its vector anopheles stephensi, with little non-target effects on predatory copepods. Environ Sci Pollut Res 2016;16:16671-85.

  187. Murugan K, Panneerselvam C, Samidoss CM, Madhiyazhagan P, Suresh U, Roni M, et al. In vivo and in vitro effectiveness of azadirachta indica-synthesized silver nanocrystals against Plasmodium berghei and Plasmodium falciparum, and their potential against malaria mosquitoes. Res Vet Sci 2016;106:14-22.

  188. Ponarulselvam S, Panneerselvam C, Murugan K, Aarthi N, Kalimuthu K, Thangamani S. Synthesis of silver nanoparticles using leaves of catharanthus roseus Linn. G. Don and their antiplasmodial activities. Asian Pac J Trop Biomed 2012;7:574.

  189. Starke CG. Leishmaniasis; 2017. Available from: http://emedicine.medscape.com/article/220298-overview [Last accessed on 12 Jul 2017]

  190. Mohapatra S. Drug resistance in leishmaniasis: newer developments. Trop Parasitol 2014;4:4–9.

  191. Ameneh S, Khadije M, Ahmad Reza T, Omid R. Inhibition of leishmania major growth by ultraviolet radiation B with silver nanoparticles in an animal model. In: Proceedings of the World Congress on Advances in Nano, Biomechanics, Robotics and Energy Research, Seoul, Korea; 2013. p. 25-8.

  192. Rossi Bergmann B, Pacienza Lima W, Marcato PD, De Conti R, Duran N. Therapeutic potential of biogenic silver nanoparticles in murine cutaneous leishmaniasis. J Nano Res 2012;20:89–97.

  193. Lima DD, Gullon B, Cardelle Cobas A, Brito LM, Rodrigues KA, Quelemes PV, et al. Chitosan-based silver nanoparticles: a study of the antibacterial, antileishmanial and cytotoxic effects. J Bioact Compat Polym 2017;4:397-410.

  194. Ahmad A, Syed F, Shah A, Khan Z, Tahir K, Khan AU, et al. Silver and gold nanoparticles from sargentodoxa cuneata: synthesis, characterization and antileishmanial activity. RSC Adv 2015;90:73793-806.

  195. Zahir AA, Chauhan IS, Bagavan A, Kamaraj C, Elango G, Shankar J, et al. Green synthesis of silver and titanium dioxide nanoparticles using Euphorbia prostrata extract shows shift from apoptosis to G0/G1 arrest followed by necrotic cell death in Leishmania donovani. Antimicrob Agents Chemother 2015;8:4782-99.

  196. Sheet F. Leishmaniasis; 2017. Available from: http://www.who.int/mediacentre/factsheets/fs375/en/.Article [Last accessed on 11 Mar 2018]

  197. Khosravi A, Sharifi I, Barati M, Zarean M, Hakimi Parizi M. Anti-leishmanial effect of nanosilver solutions on Leishmania tropica promastigotes by in vitro assay. Zahedan J Res Med Sci 2011;13:e93813.

  198. Allahverdiyev AM, Abamor ES, Bagirova M, Ustundag CB, Kaya C, Kaya F, et al. Antileishmanial effect of silver nanoparticles and their enhanced antiparasitic activity under ultraviolet light. Int J Nanomed 2011;6:2705.

  199. Hedley L, Wani RLS. Helminth infections: diagnosis and treatment; 2015. Available from: http://www.pharmaceutical-journal.com/learning/learning-article/helminth-infections-diagnosis-andtreatment/20069529.article [Last accessed on 11 Mar 2018]

  200. Rashid MM, Ferdous J, Banik S, Islam MR, Uddin AM, Robel FN. Anthelmintic activity of silver-extract nanoparticles synthesized from the combination of silver nanoparticles and M. charantia fruit extract. BMC Complementary Altern Med 2016;1:1-6.

  201. Karthik L, Kumar G, Keswani T, Bhattacharyya A, Reddy BP, Rao KB. Marine actinobacterial mediated gold nanoparticles synthesis and their antimalarial activity. Nanomedicine 2013;7:951-60.

  202. Dutta PP, Bordoloi M, Gogoi K, Roy S, Narzary B, Bhattacharyya DR, et al. Antimalarial silver and gold nanoparticles: green synthesis, characterization and in vitro study. Biomed Pharmacother 2017;91:567-80.

  203. Sazgarnia A, Taheri AR, Soudmand S, Parizi AJ, Rajabi O, Darbandi MS. Antiparasitic effects of gold nanoparticles with microwave radiation on promastigotes and amastigotes of leishmania major. Int J Hyperthermia 2013;1:79-86.

  204. Kar PK, Murmu S, Saha S, Tandon V, Acharya K. Anthelmintic efficacy of gold nanoparticles derived from a phytopathogenic fungus, Nigrospora oryzae. PloS One 2014;1:e84693.

  205. Jameii F, Dalimi Asl A, Karimi M, Ghaffarifar F. Healing effect comparison of selenium and silver nanoparticles on skin leishmanial lesions in mice. Sci J Hamadan Univ Med Sci 2015;3:217-23.

  206. Mohapatra SC, Tiwari HK, Singla M, Rathi B, Sharma A, Mahiya K, et al. Antimalarial evaluation of copper [II] nanohybrid solids: inhibition of plasmepsin II, a hemoglobin-degrading malarial aspartic protease from Plasmodium falciparum. J Biol Inorg Chem 2010;3:373-85.

  207. Dorostkar R, Ghalavand M, Nazarizadeh A, Tat M, Hashemzadeh MS. Anthelmintic effects of zinc oxide and iron oxide nanoparticles against toxocara vitulorum. Int Nano Lett 2017;2:157-64.