Logo-apb
Advanced pharmaceutical bulletin. 10(4):512-523. doi: 10.34172/apb.2020.063

Review Article

Magnetic Nanosystems as a Therapeutic Tool to Combat Pathogenic Fungi

Heba Salah Abbas 1, 2, 3, *ORCID logo, Akilandeswari Krishnan 3
1National Organization for Drug Control and Research, Cairo, Egypt.
2Scientist Under Scheme of Asian Research Training Fellowship for Developing Country (RTF-DCS), FICCI, NewDelhi, India.
3Department of Pharmaceutical Technology, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli-620024. Tamilnadu, India.
*Corresponding Author: Heba Salah Abbas, Email: Heba181179@yahoo.com

Abstract

The overuse of antibiotics is the main reason for the expansion of multidrug-resistant microorganisms, especially, pathogenic fungi, such as Candida albicans and others. Nanotechnology provides an excellent therapeutic tool for pathogenic fungi. Several reports focused on metal oxide nanoparticles, especially, iron oxide nanoparticles due to their extensive applications such as targeted drug delivery. Using biological entities for iron oxide nanoparticle synthesis attracted many concerns for being eco-friendly, and inexpensive. The fusion of biologically active substances reduced and stabilized nanoparticles. Recently, the advancement and challenges for surface engineered magnetic nanoparticles are reviewed for improving their properties and compatibility. Other metals on the surface nanoparticles can enhance their biological and antimicrobial activities against pathogenic fungi. Furthermore, conjugation of antifungal drugs to magnetic nanoparticulate increases their antifungal effect, antibiofilm properties, and reduces their undesirable effects. In this review, we discuss different routes for the synthesis of iron oxide nanoparticles, surface coating manipulation, their applications as antimicrobials, and their mode of action.

Keywords: Candida infections, Iron NPs synthesis, Magnetic nanosystems, Surface modification, Antibacterial, Antifungal mechanism

Copyright

© 2020 The Authors.
This is an Open Access article distributed under the terms of the Creative Commons Attribution (CC BY), which permits unrestricted use, distribution, and reproduction in any medium, as long as the original authors and source are cited. No permission is required from the authors or the publishers.


Introduction

Recently, the overload of fungal diseases causes 1 500 000 global deaths every year. 1 Candida species produces severe infections that may involve damage of crucial organs. 2 One hundred and fifty various species of the genus Candida were recognized including C. albicans, C. krusei , C. glabrata, C. tropicalis, C. parapsilosis , C. lusitaniae , C. dubliniensis , C. Kefir, C. guilliermondii and C. stellatoidea . They can cause human infectionsand the most invasive are infections caused byC. albicans. 3-5 C. albicans is one of the normal floras which are found in vagina, mouth, and dorsum of the tongue. The increase of candidiasis occurrence is closely related to the immunodeficiency syndrome in human. C. albicans can cause systemic infections in immunocompromised patients, such as endocarditis, and lung and brain infections. Even any change in the commensal organisms of the intestine, because of antibiotic treatments, leads to intestinal candidiasis. Infants can also be infected by vaginal candidiasis during delivery and their contact with the vagina. 6,7

In most populated countries such as Egypt, around 1 307 766 adult women suffered from vulvovaginal candidiasis in 2012. Also, candidaemia and intra-abdominal candidiasis were estimated by 4127 and 806 cases. 8 In India, high incidence of candidemia was recorded in an intensive care unit. 9 In China, Candida auris has been isolated from hospital women but, it was less virulent than C. albicans. The emergence of multidrug-resistant C. auris and its relation with high mortality is a critical issue. 10

The virulence factors ofCandida species which are responsible for pathogenicity include their effect on the host defenses by adherence, biofilm creation or/and production of proteases, phospholipases, and others that damage the host tissue. 11 Various antifungal drugs are available for the treatment of candidiasis such as amphotericin B but, it has poisonous effects. Fluconazole is safer but, certain Candida species are resistant to it. 12 The emergence of resistance against pathogenic fungi to fluconazole and amphotericin B is a major public health concern. There is an urgent demand to develop new antifungal agents.

Nanotechnology draws the attention of many researchers due to its various applications. The activities of nanoparticles largely depend on particle size. The properties of nanoparticles can change by decreasing the particle size at nanometer scale. 13-15 Green nanotechnology employs the use of biological sources such as microorganisms, plants or algae extract for the synthesis of nanomaterials. Green approaches produce safe and eco-friendly nanomaterials due to the absence of toxic substances during synthesis. 16

Magnetic nanoparticles are oneof the most important metal oxides because of their widespread applications in biotechnology and medicine. 17,18 Recently, the encapsulation of fungal drug in nanoparticle schemes offers an innovative alternative approach that promotes therapeutic efficiency and decreases the inappropriate side effects of the drugs. Limited studies were carried on the antifungal activities of biosynthesized Iron oxide nanoparticles. The antifungal activity of biosynthesized iron oxide nanoparticles was previously investigated. 19 Iron oxide nanoparticles cause inhibition for growth and spore germination of Trichothecium roseum , Cladosporium herbarum , Penicillium chrysogenum , Alternaria alternata and Aspergillus niger . The continual resistance of microorganisms led to advancement of chitosan coated iron oxide nanoparticles as new antimicrobial agents against Escherichia coli , Bacillus subtilis , C. albicans , A. niger and, Fusarium solani. 20 Our study aims to discuss routes for synthesis of iron oxide nanoparticles, surface coating manipulation and, their potential use as new antifungal agents.


Methods for Synthesis of iron oxide nanoparticles

Physical methods

Ironoxide nanoparticlescan be synthesized via various techniques such as chemical, physical, and biological techniques (). There are different methods for physical synthesis of Iron oxide nanoparticles such as pyrolysis, laser ablation, etc.

apb-10-512-g001
Figure 1. Flow chart for preparation methods of Iron oxide nanoparticles.

Laser ablation method depends on the solvent used whether it is organic, or inorganic solvent such as ethanol, or acetone. In general, ethanol and acetone are better than organic solvents because organic solvents can elaborate various by-products, with different physical and chemical characters, which show influence on nanoparticles stability. 21 Using Polymers during the synthesis of iron oxide nanoparticles can control their size and distribution. This stabilized iron oxide nanoparticles showed good antimicrobial characterstics. 22

In spray pyrolysis or gas/aerosol method , ferric salt solution and a reducing agent sprayed and the aerosol solute condensed during the solvent evaporation. The yield percentage is very low and the equipment for this method is very expensive. 23,24 The most disadvantage of this method is the uncontrollable size of nanoparticle in nanometer range. 25

Also, Kang and Rhee have studied the impact of pressure (60 torr) and 800°C temperature on ultrasonic spray pyrolysis by using acetate and nitrate solutions for the synthesis of manganese, nickel, and copper oxide. The products were hollow shaped submicron particles with large crystalline size (>40 nm) and nanoparticles with small crystalline size (<10 nm). 26 In other study, Ozcelik and Ergun elucidated that the crystallinity of the spherical iron oxide increased by increasing temperature to 1100°C. 27

Chemical methods

Various techniques are documented for the chemical synthesis of nanoparticles such as coprecipitation, microemulsion, hydrothermal, thermal decomposition, and sonochemical methods. They are categorized by their simplicity, low-cost, and high yield of nanoparticles with controlled morphology.

Incoprecipitation , iron oxide nanoparticles are synthesized by adding base into ferric chloride solution followed by precipitation black coloured magnetite. Magnetite precipitates in alkali conditions (pH 9-14) and in the absence of oxygen. Otherwise, it is oxidized into hydroxide form as in the subsequent equation: -

Fe3O4 + 0:25O2 + 4:5H2O → 3Fe (OH) 3+

The bubbling of nitrogen gas during the process of synthesis protects iron oxide nanoparticles from oxidation and decreases their size. Also, the coating of nanoparticles by using organic and inorganic molecules prevents their agglomeration and oxidation.

The kind of salt precursor, ferrous/ferric ratio (1:2), pH, ionic strength, temperature, and the bubbling of nitrogen gas can influence the morphology of iron oxide nanoparticles. 23,28

Also, Nazari et al used wool fabrics and butane tetracarboxylic as a stabilizer for iron oxide nanoparticles to get better results as antifungals against C. albicans . 29

Thehydrothermal method requires high pressure (>2000 psi) and temperature (>200°C). The reaction depends on hydrolyzing the metal salt by water in autoclave or reactor. However, this method takes long time and elevated temperature for synthesis and this causes effect on the size and morphology of metal oxide nanoparticulate.25, 30-32

In microemulsion method (two phases method), the nano-water droplet disperses in oil and is stabilized by surfactant. The surfactant type may be cationic, anionic, or none-anionic form. The core advantage of this method is the production of diverse nanoparticles by changing reaction conditions like introducing an oil phase or changing the quantity of surfactant. 33 However, the disadvantages are: low temperature, large amount of oil that make large-scale production difficult, and the effect of residual surfactant on nanoparticles properties. 34-36

In thermal decomposition method , iron salt precursors decompose thermally without oxygen and produce a high yield of Iron oxide nanoparticles. However, the product is mixture of nano-iron oxide phases with crystal defects, and also, its hydrophobic nature needs additional stages to be compatible with hydrophilic surface. 37,38 During the thermal decomposition process, Unni et al synthesized a single nano-iron oxide phase with limited defect by addition of oxygen. 39

In the sonochemical method , iron precursor such as ferric chloride hexahydrate is decomposed by high intensity of ultrasonication then polymers are added for capping and stopping nanoparticles growth. Cavitation can occur due to ultrasonic irradiation, with a consequent increase in temperature to reach 5000°C and of pressure to exceed 1800 kPa, causing anomalous chemical reactions (Table 1). 39,40

Table 1. Physical and chemical preparation methods for iron oxide nanoparticles, types of particles, morphology, advantages, and disadvantages of different methods
Methods Nanoparticles Morphology Types of
Particles
Advantage Disadvantage References
Physical-laser ablation methodSpherical, 20-100 nmMaghemite- HematiteStable with a narrow size distribution only in Polymeric solutionUncontrolled size in water solution 22
Physical-spray pyrolysisSpherical, 70-675 nmHematiteUniform morphology Crystallinity increases by High temperature (1100oC) 27
Chemical-coprecipitation Nanocubes (7.8 ± 0.05 nm) and nanorod (6.3 ± 0.2 nm)Magnetite Small sized nanoparticles,
Simple reaction conditions
28
Chemical-hydrothermal Spherical (15.6±4.0 nm) or Rhombic (27.4±7.0 nm)MaghemiteSmall sized nanoparticles High pressure and temperature requirements.
It easily affected by precursor concentration
32
Chemical-microemulsonSpherical, <10 nmMagnetite or MaghemiteDiverse nanoparticles 33
Chemical-thermal decomposition SphericalMixed phases High yield Poor and crystal defects.
Hydrophobic nature.
39
Chemical-sonochemical Spherical >19 nmHematiteSmall sizeHigh temperature and pressure 41

Biological methods

Biological methods have more advantages over the conventional chemical and physical methods like being non-polluting and eco-friendly. Besides, they have low cost of synthesis since the biological active material acts as reducing and capping agent and produces high yield of small sized nanoparticles (). The biological synthesis method aid in iron oxide nanoparticle coating compared to chemical synthesis method. 42

apb-10-512-g002
Figure 2. Mechanism of iron oxide nanoparticles biosynthesis.

Many research papers elucidated the biosynthesis of iron oxide nanoparticles (Table 2) with different sizes and shapes from plant extracts such as Hordeum vulgare and Rumex acetosa extracts. H. vulgare contains high amounts of reducing compounds compared toR. acetosa extract. However, iron oxide nanoparticles produced by H. vulgare were aggregated and unstable. The aggregation and instability problem can be resolved by organic acids in the form of citrate, malate, and oxalate coating. The total protein content and antioxidants properties were similar for the two plant extracts. The stability of iron oxide nanoparticle byR. acetosa extract were because of pH 3.7 compared to instability of iron oxide nanoparticle byH. vulgare extract which has pH 5.8. 43

Table 2. Biological preparation methods for iron oxide nanoparticles, types of particles, morphology, advantages, and disadvantages of different methods
Biological Methods Nanoparticles Morphology Types of
Particles
Advantage Disadvantage References
Plant - Hordeum vulgare Spherical -30 nmMixed iron oxidation statesEco-friendly Instability and aggregation of nanoparticles with time 42
Plant - Rumax acetosa Amorphous -40 nmMixed iron oxidation states Eco-friendly
Highly Stable
- 42
Plant - Amaranthus spinosus Spherical
91nm
rhombohedral crystalline structure of hematite Eco-friendly
Stable
- 43
Plant - green tea Spherical
70-80 nm
Maghemite, magnetite and iron hydroxidesEco-friendly- 44
Plant - sorghum bran Amorphous
50 nm
Lack distinct diffraction peaksEco-friendlyAgglomeration and irregular clusters 45
Plant - pomegranate Spherical
10-30 nm
--No agglomeration- 46
Brown Algae- Sargassum muticum Spherical-18 ± 4Cubic formEco-friendly-stable -small size- 49
Green Algae - Chlorococcum sp. Spherical
50 nm
-Eco-friendly- highly stable - 50
Fungi - Aspergillus japonicus Cubic 60-70 nmMagnetite and maghemite Stable - 51
Fungi - Fusarium oxysporum and Verticillium sp Quasi-spherical
20-50 nm
Magnetite and maghemiteStable- 52
Fungi - Verticillium sp Cubo-octahedrally 100-400 nmMagnetite and maghemiteStable 52
Bacteria - Actinobacter sp. Spherical 19 nmMaghemiteStable- 53

Also, Amaranthus spinosus water leaf extract is added to ferric chloride for the synthesis of spherical iron oxide nanoparticles. The presence of amaranthine and phenolic compounds in this aqueous extract allows the reduction process and capping of iron oxide nanoparticles. 44 Spherical IONs can be also biosynthesized by using ferric sulphate as precursor and green tea extracts as reducing agent. Characteristic UV peaks are observed at 205 and 272 nm and this is an indication for presence of polyphenols and caffeine in green tea extract. Polyphenols reduce iron salts and is capping it. The diameter of these nanoparticles was 70-80 nm. In general, the reduction potential of polyphenols/caffeine was in 0.3-0.8 V and iron reduction potential was -0.44 V. 45 Also, adding ferric chloride solution into sorghum bran extract leads to formation of amorphous iron oxide nanoparticles with an average diameter of 50 nm. The polyphenols in sorghum extract stabilizes the biosynthesized iron oxide nanoparticles. 46

Polyphenols are essential components in the reduction process of iron salts into zerovalent iron oxide nanoparticles because of its antioxidant property. 46

The possible mechanism for biosynthesis of iron oxide nanoparticles is explained 19 as follows:

apb-10-512-g008
.

The antifungal features of iron oxide nanoparticles produced by a phenolic form of tannic acid were well studied, which will control fungal diseases. 19 The use of anhydrous ferric chloride and ferrous chloride hydrate mixtures as a precursor with 6% tangerine peels extract can synthesize spherical iron oxide nanoparticles with an average diameter 50 nm. Increasing the concentration of extract causes sever aggregation of nanoparticles. 47 Even extracts of several tree leaves such as almond, apricot, avocado, cherry, eucalyptus, kiwi, lemon, mandarin, medlar, mulberry, green tea, black tea oak, olive, orange, passion fruit, peach, pear, pine, pomegranate, plum, quince, raspberry, strawberry, vine, and walnut are investigated for reduction of iron(III) chloride hexahydrate to zero valent iron nanoparticles oxide (d = 10-30 nm). According to their antioxidant activity, green tea, pomegranate and black tea water extracts showed higher antioxidant activity compared to other tree leaves because they are rich with phenolic content. 47 Moreover, using a polysaccharide template as Chitosan for biosynthesis of spherical -shaped iron oxide nanoparticles is recorded which aided the coating by sand. 42 Chitosan can change the morphology of iron oxide nanoparticles from rod like, flower like and, cubo-octahedral structures into rice-seed-like, quasi-spherical, and cubic structures, respectively. 49

Other reports elucidated the mechanism of iron oxide nanoparticles production by sulphated polysaccharide of brown see weedsSargassum muticum extract. 50 Also soil microalgae Chlorococcum sp. can synthesize spherical nano-iron extracellularly and intracellularly. Glycoprotein and polysaccharide mediated the synthesis and stabilization of nanoiron. 51

On the other hand, fungal protein mediated the biosynthesis of iron oxide nanoparticles. Cationic protein content of Aspergillus japonicus isolate AJP01, Fusarium oxysporum and Verticillium sp. can hydrolyse anionic iron cyanide complexes and produce iron oxide nanoparticles. Nanoparticulate magnetite has size range of 50-60 nm for A. japonicus and 20-50 nm for F. oxysporum and Verticillium sp. 52,53 The protein analysis elucidated the presence of two proteins with molecular weight 55 and 13 kDa which are responsible for hydrolysing mixture of iron cyanide complexes and capping of nanoparticulate magnetite. 53 Also, Iron reductase in bacteria may play role in reduction of iron salt during formation of bacterial maghemite nanoparticles by Actinobacter sp. A protein of 55 kD was observed and other new proteins were induced during the biosynthesis process. These new proteins are responsible for capping and stabilization of nanoparticles. 54


Properties of iron oxide nanoparticles

There are three types of iron oxide nanoparticles; magnetite, maghemite and hematite. The hematite is red in colour if finely divided or black-grey in colour if crystallized. Magnetite also is black in colour and has strong magnetism. Maghemite is an oxidized metastable product of iron oxide. The instability problem of maghemite at high temperature can be resolved by doping it with other metals. Maghemites can loss its magnetism by irreversible conversion into hematite at around 400°C. 55-58 Small size of maghemites (<10 nm) is super paramagnetic at ordinary temperature. The magnetic properties of iron oxide nanoparticles are influenced by surface effects. The magnetic properties are lost faster by increasing temperature. Chemical method for surface modification of iron oxide nanoparticles influences their coercivity. The size, nanostructure surface treatments and, method of preparation can change the magnetic properties. 58-62

Certain sizes, shapes, surface characteristics and magnetic properties of iron oxide nanoparticles are depending upon the used application. The application of iron oxide nanoparticles in biology and medical diagnosis demands the stability of nanoparticles during the physiological conditions. 63,64 The small dimension of nanoparticles, charge and surface chemistry have influence on stability of colloidal magnetic fluid. Magnetite and maghemite with external magnetic stimuli allow drug delivery and permit low dose administration. 64,65 Moreover, functionalization of nanoparticles increases therapeutic efficiency. 65


Surface modification of magnetic nanoparticles

Iron oxide nanoparticles may be insoluble and non-biocompatible; Therefore, the surface should manipulate to improve biocompatibility. 66,67 In general, there are several reasons for surface modification of iron oxide nanoparticles; improvement of the dispersion, surface activity, physicochemical, and mechanical properties can improve the biocompatibility of iron oxide nanoparticles. 67 There are different shapes of magnetic nanocomposite as in . 68,69

apb-10-512-g003
Figure 3. Morphology of magnetic nanocomposites.

Several strategies are used for functionalizing iron oxide nanoparticlesfor the stability of colloidal suspension or other desired applications. 70 Iron oxide nanoparticles can be covered by a shell of organic (surfactants or polymers) or inorganic (carbon or silica) or bioactive molecules as in . 23

apb-10-512-g004
Figure 4. The main shells for manipulating Iron oxide nanoparticle (grey circle).

The polymers can be synthetic as in the forms of polyethylene glycol, polyvinylpyrrolidone, and polyvinyl alcohol or natural as in the form of chitosan. 23,68,71 The advantage of hydrophilic uncharged polyethylene glycol, when used in the coating of iron oxide nanoparticles, is that it cannot be recognized by the immune system, and this helps to stay in the blood circulation for a long time and gathering in the target organ. 71,72 In the case of using the hydrophilic polyvinylpyrrolidone, and polyvinyl alcohols which have hydrogel structures so it can be linked with iron oxide nanoparticles by hydrogen bonds, and interactions between polymer and surface can be increased which prevent nanoparticles aggregations. 72

However, a natural polymer such as chitosan has a positive charge that drives chitosan carriers to negatively charged cell membranes besides their mucoadhesive characteristics, which cause their retention on target cells. 73 The magnetic and thermal properties of iron oxide nanoparticles cannot be changed by chitosan coating. It was hypothesized that the electrostatic repulsion between the negative potential surface of iron oxide nanoparticles and bacteria lowers the antimicrobial activity compared to the positive potential surface of iron oxide nanoparticles. 74 However, the partial protonation of amino groups in chitosan coating reduces its water solubility. To overcome such problem, using O-carboxymethyl chitosan or carboxymethyl starch chitosan can be used via some chemical changes to get water solubilization. 23,70,75,76 Also, sodium alginate as polysaccharide used for grafting magnetic nanospheres and encapsulated by cisplatin to control release the cisplatin dug. 77

The modification of the shell surface of iron oxide nanoparticles by using a hydrophilic group is one of the most suitable methods for desired applications such as magnetic targeting delivery and hydrothermal cancer therapy.For example,Fe3O4@ dopamine was used as enzyme mimetic for the detection of bacteria. 78 Moreover, Iron oxide nanoparticles functionalized with amine groups using (3-aminopropyl) trimethoxysilane. The conjugation of amino with doxorubicin is followed by bonding with bi-functional polyethylene glycol and then folic acid for targeting the tumor. The hydrophobic core is DOX conjugated with iron oxide nanoparticles and polyethylene glycol-OCH3/Folic acid, which acts as a shell nanocarriers. Magnetic core aid not only targets the drug for carrying to tumor cells but can also be used for magnetic resonance imaging. 79

Non-polymer organic molecules such as alkanesulphonic or alkanephosphonic acids, oleic, lauric, dodecylyphosphonic, hexadecylphosphonic acids are used for stabilization of iron oxide nanoparticles in organic solvent. 80,81 However, a long hydrocarbon chain causes the hydrophobic nature of nanoparticles that hinders in vivo applications. 82

Inorganic coating materials like silicon dioxide or carbon are favored in biological labeling or optical bioimaging or in increasing the antioxidant properties. Silicon dioxides coating of nanoparticles maintain the stability of nanoparticles in acidic medium and reduce the toxicity of iron oxide nanoparticles. 83-85 Also, the carbon coating of iron oxide nanoparticles prevents iron nanoparticles from oxidation besides, the diverse properties of carbon such as stability at different temperatures, good electrical conductivity, and solubility. 71 The metal coating of nanoparticles prevents the low reactivity of nanoparticles. 68 Positively charged silver coating allows the conjugation of different antibiotics. 86 The possible combination between metal oxides creates intrinsic magnetic properties. The selection of coating depends on the purpose of the application. For example, zinc oxide nanoparticle was chosen as a suitable compound for anticancer nano-composite using trisodium citrate as a linker for conjugation of Fe3O4 with ZnO. The hypothesis for anticancer activity was the reactive oxygen species, which cause the selective cytotoxicity of ZnO and exhaust the activity of scavenging of cancerous cells. Therefore, it promotes the cytotoxicity of iron oxide nanoparticles against cancerous cells. 87 Moreover, ZnO nanoparticles have the capability of inhibiting pathogenic bacteria, yeast, and filamentous fungi. 88

Bioactive molecules such as lipids, peptides, and proteins can be coated with iron oxide nanoparticles for improving their stability and magnetic properties. 72,76


Antibacterial and antifungal iron oxidenanosystems

Biocidal activity of metals

Since ancient times, the toxicity of metals is known to bacteria, fungi, and has been used as antimicrobial agents. The possible mechanism is not well elucidated. In general, the biocidal activity of metals depends on the potential of metal reduction and selectivity. 89-91 The metal toxicity mechanisms () explained as follow:

apb-10-512-g005
Figure 5. Possible mechanisms behind biocidal activity of metals

The potential of metal reduction acts as a cofactor for activating cell enzymes and generation of reactive oxygen species (ROS) that can induce oxidative stress resulting and subsequently in proteins, lipids, and DNA damage. Besides, the excess of ROS induces proinflammatory signals, which cause programmed cell death. 91,92 The main principle for metal toxicity is the production of reduced forms of oxygen molecules such as hydrogen peroxide and superoxide during aerobic respiration. Hydrogen peroxide can react with metals like iron and produces hydroxide and hydroxide radicals (Fenton reactions). The hydroxide radicals can react with biological molecules such as amino carbon compounds and form carbon-protein radicals or with unsaturated fatty acids and form lipid radicals. Some metals can form protein disulfides by binding with sulfur and causes depletion of glutathione reservoirs. Besides, this mechanism depends upon the selectivity of metal donors, in which the metal ions bind with another atom such as nitrogen, oxygen, and sulfur. Therefore, metal ions or its complexes can replace the original biomolecules metals and causes cell dysfunction. Metals can cause inactivation of enzymes and promote Fe-S clusters. 89,90 Other mechanisms depend upon cell membranes or intracellular region. For instance, bacterial membranes have highly electronegative macromolecules that are the site for adsorption for metals. Therefore, cell membranes are the first barrier that damaged by metal ions that permit subsequent intracellular uptake and causes bactericidal toxicity. 93

Antimicrobial activity of metal nanoparticles

Metal nanoparticles should be stronger antimicrobials than metals because of their nanoscale size, and their unique physical and chemical properties. Metal nanoparticles can incorporate directly inside the cell by endocytosis. Hence, the uptake of ions through the cell increases in the form of ionic species and released within the cell. This process is called a Trojan-horse mechanism. Besides the oxidative stress occurs inside the cell. 94 A probable mechanism for antimicrobial effect of metal nanoparticles is showed in .

apb-10-512-g006
Figure 6. A probable mechanism for antimicrobial effect of metal nanoparticles :1- endocytosis, 2- attachment of membrane surface, 3-free radical formation, and 4- release of metal ions.

Coated and non-coated iron oxide nanoparticles as therapeutic tools to combat pathogenic microorganisms

Iron oxide nanoparticles adhere to bacterial cell membranes and cause membrane depolarization and loss of membrane integrity. Besides, damage of deoxyribonucleic acid and protein via generation of ROS occurs with lipid peroxidation. 95 The presence of metal ions inside the cell causes cell imbalance and affects the protein harmony. 96 Rod-shaped iron oxide nanoparticles synthesized by water extract of Spirulina platensis penetrate the cell membrane and cause deformation for the morphology of multidrug-resistant Helicobacter pylori (). 97 As a result of continuous leakage of intracellular content and shrinkage of the cell membrane, the death of bacteria occurs.

apb-10-512-g007
Figure 7. Transmission electron microscope Images of rode shaped iron oxide nanoparticles synthesized by water extract of Spirulina platensis (A) and deformation of multidrug resistance Helicobacter pylori after treatment with MIC50 of iron oxide nanoparticles (red circle) (B).

Carboxylate functionalized iron oxide nanoparticles penetrate the biofilm of bacteria and reduce their growth. 98 Also, gold-coated iron oxide nanoparticles can adhere to the bacterial protein by disulfide bonds and influence the on bacteria metabolism by increasing the permeability of cell membranes causing damage to the bacterial cell wall. Changes in the morphology of Pseudomonas aeruginosa can occur due to the interaction of gold-coated iron oxide nanoparticles with protein F, which has the main role in the resistance of bacteria against antibiotics. 99 Magnetic iron oxide nanoparticles can catch gram-positive and gram-negative bacteria because of the presence of protein F in both. 100

Metals can be incorporate on polymer surface or impregnated into the matrix. These materials possess both antibacterial and antifungal activities. The antimicrobial mechanism of polymer@ metal nanocomposite depends on metal nanoparticles and free metal ion received from metal nanoparticles. Several reports recorded the importance of released metal ions in the antimicrobial activity of polymer@ metals nanocomposite. 101 Microorganisms can form a biofilm to adhere to the biomaterial surfaces and protect itself from antibiotics and host defence mechanisms. The biofilm growth can be reduced in the presence of a polymer brush combined with a high concentration of iron oxide nanoparticles. 102

Combination with metal nanoparticles is considered as an alternative approach to overcome the resistance of microorganisms to the antibiotics. 103 Therefore, loading nystatin antifungal drugs on chitosan-coated iron oxide nanoparticles showed a comparable enhancement in fungal activity against C. albicans . Besides, it showed better antimicrobial activity against P. aeruginosa and Escherichia coli than Staphylococcus aureus . 104 Also, the conjugation of two polyene antibiotics such as amphotericin B and nystatin to magnetic nanoparticles increase the antifungal/antibiofilm activity against clinical isolates of Candida species. The mechanism of antifungal/antibiofilm activity has been investigated as the cause for inactivation of catalase and imbalance of oxidation-reduction that inhibits Candida growth. Hemolytic activity of polyene antibiotics against human red blood cells decreased after magnetic nanoparticle conjugation. 105 A group of researchers prepared two magnetic nanocomposites @ silver nanoparticles by using a polyacrylate linker. Nanocomposites possess significant antibacterial and antifungal activity against different bacteria strains and Candida species. 105 In that concern, Prucek et al thermally synthesized iron oxide nanoparticles conjugated with silver nanoparticles with good antimicrobial activities that can be used in biomedical applications as disinfectants. 106 Also, Wilczewska et al investigated that the conjugation of magnetic nanocarriers with metallocarbonyl complexes showed good antifungal activity against C. albicans . 107


Conclusion and Future Prospects

The Surface coating of Iron oxide nanoparticles not only decreases the cytotoxicity of iron oxide nanoparticles but also increases the stability and efficiency of antifungal and anticancer properties of nanoparticles. The coating of Iron oxide nanoparticles with metal or other metal oxide nanoparticles may even cause a revolution in the therapeutic world.


Ethical Issues

Not applicable.


Conflict of Interest

Authors declare no conflict of interest in this study.


Acknowledgments

Authors thankfully acknowledge the financial support provided by FICCI, DST, New Delhi, Government of India [DCS/2018/000048], Asian Research Training Fellowship Scheme for Developing Country Scientist (RTF-DCS), and Bharathidasan Institute of Technology, Anna University, Tiruchirappalli-620024.Tamilnadu, India, and the Microbiology Department, National Organization for Drug Control and Research, Giza, Egypt.


References

  1. Global Action Fund for Fungal Infections. 2015. ‘95–95 by 2025’ Improving outcomes for patients with fungal infections across the world: a roadmap for the next decade. May 2015. https://www.gaffi.org/roadmap/.
  2. Eckert LO, Hawes SE, Stevens CE, Koutsky LA, Eschenbach DA, Holmes KK. Vulvovaginal candidiasis: clinical manifestations, risk factors, management algorithm. Obstet Gynecol 1998; 92(5):757-65. doi: 10.1016/s0029-7844(98)00264-6 [Crossref]
  3. James WD, Berger TG, Elston DM; Richard B Odom RB. Andrews’ Diseases of the Skin: Clinical Dermatology. Philadelphia: Saunders Elsevier; 2006. p. 308-11.
  4. Pfaller MA, Diekema DJ. Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev 2007; 20(1):133-63. doi: 10.1128/cmr.00029-06 [Crossref]
  5. Johnson EM. Rare and emerging Candida species. Curr Fungal Infect Rep 2009; 3(3):152-9. doi: 10.1007/s12281-009-0020-z [Crossref]
  6. Delaloye J, Calandra T. Invasive candidiasis as a cause of sepsis in the critically ill patient. Virulence 2014; 5(1):161-9. doi: 10.4161/viru.26187 [Crossref]
  7. Dowd FJ. Candida Albicans Infections. Reference Module in Biomedical Sciences. 2014.
  8. Zaki SM, Denning DW. Serious fungal infections in Egypt. Eur J Clin Microbiol Infect Dis 2017; 36(6):971-4. doi: 10.1007/s10096-017-2929-4 [Crossref]
  9. Chakrabarti A, Sood P, Rudramurthy SM, Chen S, Kaur H, Capoor M. Incidence, characteristics and outcome of ICU-acquired candidemia in India. Intensive Care Med 2015; 41(2):285-95. doi: 10.1007/s00134-014-3603-2 [Crossref]
  10. Wang X, Bing J, Zheng Q, Zhang F, Liu J, Yue H. The first isolate of Candida auris in China: clinical and biological aspects. Emerg Microbes Infect 2018; 7(1):93. doi: 10.1038/s41426-018-0095-0 [Crossref]
  11. Silva S, Negri M, Henriques M, Oliveira R, Williams DW, Azeredo J. Candida glabrata, Candida parapsilosis and Candida tropicalis: biology, epidemiology, pathogenicity and antifungal resistance. FEMS Microbiol Rev 2012; 36(2):288-305. doi: 10.1111/j.1574-6976.2011.00278.x [Crossref]
  12. Smeekens SP, Ng A, Kumar V, Johnson MD, Plantinga TS, van Diemen C. Functional genomics identifies type I interferon pathway as central for host defense against Candida albicans. Nat Commun 2013; 4:1342. doi: 10.1038/ncomms2343 [Crossref]
  13. Lewis K, Klibanov AM. Surpassing nature: rational design of sterile-surface materials. Trends Biotechnol 2005; 23(7):343-8. doi: 10.1016/j.tibtech.2005.05.004 [Crossref]
  14. Rosi NL, Mirkin CA. Nanostructures in biodiagnostics. Chem Rev 2005; 105(4):1547-62. doi: 10.1021/cr030067f [Crossref]
  15. 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 Nanomedicine 2012; 7:3527-35. doi: 10.2147/ijn.s29020 [Crossref]
  16. Nasrollahzadeh M, Sajjadi M, Sajadi SM, Issaabadi Z. Chapter 5 - Green Nanotechnology. In: Nasrollahzadeh M, Sajadi SM, Sajjadi M, Issaabadi Z, Atarod M, eds. Interface Science and Technology. Elsevier; 2019. p. 145-98. 10.1016/B978-0-12-813586-0.00005-5
  17. Tartaj P, Morales MP, Gonzalez-Carreño T, Veintemillas-Verdaguer S, Bomati-Miguel O, Roca Roca, AG AG, Serna CJ. Biomedical Applications of Magnetic Nanoparticles. Reference Module in Materials Science and Materials Engineering 2016:1-7.
  18. Pankhurst QA, Connolly J, Jones SK, Dobson J. Applications of magnetic nanoparticles in biomedicine. J Phys Appl Phys 2003; 36(13):R167-R81. doi: 10.1088/0022-3727/36/13/201 [Crossref]
  19. Parveen S, Wani AH, Shah MA, Devi HS, Bhat MY, Koka JA. Preparation, characterization and antifungal activity of iron oxide nanoparticles. Microb Pathog 2018; 115:287-92. doi: 10.1016/j.micpath.2017.12.068 [Crossref]
  20. Nehra P, Chauhan RP, Garg N, Verma K. Antibacterial and antifungal activity of chitosan coated iron oxide nanoparticles. Br J Biomed Sci 2018; 75(1):13-8. doi: 10.1080/09674845.2017.1347362 [Crossref]
  21. Amendola V, Meneghetti M. What controls the composition and the structure of nanomaterials generated by laser ablation in liquid solution?. Phys Chem Chem Phys 2013; 15(9):3027-46. doi: 10.1039/c2cp42895d [Crossref]
  22. Fazio E, Santoro M, Lentini G, Franco D, Guglielmino SPP, Neri F. Iron oxide nanoparticles prepared by laser ablation: synthesis, structural properties and antimicrobial activity. Colloids Surf A Physicochem Eng Asp 2016; 490:98-103. doi: 10.1016/j.colsurfa.2015.11.034 [Crossref]
  23. Arias LS, Pessan JP, Vieira APM, Lima TMT, Delbem ACB, Monteiro DR. Iron oxide nanoparticles for biomedical applications: a perspective on synthesis, drugs, antimicrobial activity, and toxicity. Antibiotics (Basel) 2018; 7(2). doi: 10.3390/antibiotics7020046 [Crossref]
  24. González-Carreño T, Morales MP, Gracia M, Serna CJ. Preparation of uniform γ-Fe2O3 particles with nanometer size by spray pyrolysis. Mater Lett 1993; 18(3):151-5. doi: 10.1016/0167-577x(93)90116-f [Crossref]
  25. Hasany SF, Ahmed I, Rajan J, Rehman A. Systematic review of the preparation techniques of iron oxide magnetic nanoparticles. J Nanosci Nanotechnol 2012; 2(6):148-58. doi: 10.5923/j.nn.20120206.01 [Crossref]
  26. Kang JK, Rhee SW. Chemical vapor deposition of nickel oxide films from Ni(C5H5)2/O2. Thin Solid Films 2001; 391(1):57-61. doi: 10.1016/s0040-6090(01)00962-2 [Crossref]
  27. Ozcelik BK, Ergun C. Synthesis and characterization of iron oxide particles using spray pyrolysis technique. Ceram Int 2015; 41(2 Pt A):1994-2005. doi: 10.1016/j.ceramint.2014.09.103 [Crossref]
  28. Khalil MI. Co-precipitation in aqueous solution synthesis of magnetite nanoparticles using iron (III) salts as precursors. Arab J Chem 2015; 8(2):279-84. doi: 10.1016/j.arabjc.2015.02.008 [Crossref]
  29. Nazari A, Shishehbor MR, Poorhashemi SM. Enhanced magnetic and antifungal characteristics on wool with Fe3O4 nanoparticles and BTCA: a facile synthesis and RSM optimization. J Text Inst 2016; 107(12):1617-31. doi: 10.1080/00405000.2015.1131439 [Crossref]
  30. Capek I. Preparation of metal nanoparticles in water-in-oil (w/o) microemulsions. Adv Colloid Interface Sci 2004; 110(1-2):49-74. doi: 10.1016/j.cis.2004.02.003 [Crossref]
  31. Hao Y, Teja AS. Continuous hydrothermal crystallization of α–Fe2O3 and Co3O4 nanoparticles. J Mater Res 2003; 18(2):415-22. doi: 10.1557/jmr.2003.0053 [Crossref]
  32. Xu C, Teja AS. Continuous hydrothermal synthesis of iron oxide and PVA-protected iron oxide nanoparticles. J Supercrit Fluids 2008; 44(1):85-91. doi: 10.1016/j.supflu.2007.09.033 [Crossref]
  33. Chin AB, Yaacob II. Synthesis and characterization of magnetic iron oxide nanoparticles via w/o microemulsion and Massart’s procedure. J Mater Process Technol 2007; 191(1-3):235-7. doi: 10.1016/j.jmatprotec.2007.03.011 [Crossref]
  34. Cuenya BR. Synthesis and catalytic properties of metal nanoparticles: size, shape, support, composition, and oxidation state effects. Thin Solid Films 2010; 518(12):3127-50. doi: 10.1016/j.tsf.2010.01.018 [Crossref]
  35. Yin Y, Li ZY, Zhong Z, Gates B, Xia Y, Venkateswaran S. Synthesis and characterization of stable aqueous dispersions of silver nanoparticles through the Tollens process. J Mater Chem 2002; 12(3):522-7. doi: 10.1039/b107469e [Crossref]
  36. Tartaj P, De Jonghe LC. Preparation of nanospherical amorphous zircon powders by a microemulsion-mediated process. J Mater Chem 2000; 10(12):2786-90. doi: 10.1039/b002720k [Crossref]
  37. Ali A, Zafar H, Zia M, Ul Haq I, Phull AR, Ali JS. Synthesis, characterization, applications, and challenges of iron oxide nanoparticles. Nanotechnol Sci Appl 2016; 9:49-67. doi: 10.2147/nsa.s99986 [Crossref]
  38. Unni M, Uhl AM, Savliwala S, Savitzky BH, Dhavalikar R, Garraud N. Thermal decomposition synthesis of iron oxide nanoparticles with diminished magnetic dead layer by controlled addition of oxygen. ACS Nano 2017; 11(2):2284-303. doi: 10.1021/acsnano.7b00609 [Crossref]
  39. Wang XK, Chen GH, Guo WL. Sonochemical degradation kinetics of methyl violet in aqueous solutions. Molecules 2003; 8(1):40-4. doi: 10.3390/80100040 [Crossref]
  40. Mason TJ, Lorimer JP. Applied Sonochemistry: Uses of Power Ultrasound in Chemistry and Processing. Weinheim: Wiley‐VCH; 2002.
  41. Hassanjani-Roshan A, Vaezi MR, Shokuhfar A, Rajabali Z. Synthesis of iron oxide nanoparticles via sonochemical method and their characterization. Particuology 2011; 9(1):95-9. doi: 10.1016/j.partic.2010.05.013 [Crossref]
  42. Rasheed R, Meera V. Synthesis of Iron Oxide Nanoparticles Coated Sand by Biological Method and Chemical Method. Procedia Technol 2016; 24:210-6. doi: 10.1016/j.protcy.2016.05.029 [Crossref]
  43. Makarov VV, Makarova SS, Love AJ, Sinitsyna OV, Dudnik AO, Yaminsky IV. Biosynthesis of stable iron oxide nanoparticles in aqueous extracts of Hordeum vulgare and Rumex acetosa plants. Langmuir 2014; 30(20):5982-8. doi: 10.1021/la5011924 [Crossref]
  44. Muthukumar H, Matheswaran M. Amaranthus spinosus leaf extract mediated FeO nanoparticles: physicochemical traits, photocatalytic and antioxidant activity. ACS Sustain Chem Eng 2015; 3(12):3149-56. doi: 10.1021/acssuschemeng.5b00722 [Crossref]
  45. Huang L, Luo F, Chen Z, Megharaj M, Naidu R. Green synthesized conditions impacting on the reactivity of Fe NPs for the degradation of malachite green. Spectrochim Acta A Mol Biomol Spectrosc 2015; 137:154-9. doi: 10.1016/j.saa.2014.08.116 [Crossref]
  46. Njagi EC, Huang H, Stafford L, Genuino H, Galindo HM, Collins JB. Biosynthesis of iron and silver nanoparticles at room temperature using aqueous sorghum bran extracts. Langmuir 2011; 27(1):264-71. doi: 10.1021/la103190n [Crossref]
  47. Machado S, Pinto SL, Grosso JP, Nouws HP, Albergaria JT, Delerue-Matos C. Green production of zero-valent iron nanoparticles using tree leaf extracts. Sci Total Environ 2013; 445-446:1-8. doi: 10.1016/j.scitotenv.2012.12.033 [Crossref]
  48. Ehrampoush MH, Miria M, Salmani MH, Mahvi AH. Cadmium removal from aqueous solution by green synthesis iron oxide nanoparticles with tangerine peel extract. J Environ Health Sci Eng 2015; 13:84. doi: 10.1186/s40201-015-0237-4 [Crossref]
  49. Vasylkiv O, Bezdorozhev O, Sakka Y. Synthesis of iron oxide nanoparticles with different morphologies by precipitation method with and without chitosan addition. J Ceram Soc Jpn 2016; 124(4):489-94. doi: 10.2109/jcersj2.15288 [Crossref]
  50. Mahdavi M, Namvar F, Ahmad MB, Mohamad R. Green biosynthesis and characterization of magnetic iron oxide (Fe₃O₄) nanoparticles using seaweed (Sargassum muticum) aqueous extract. Molecules 2013; 18(5):5954-64. doi: 10.3390/molecules18055954 [Crossref]
  51. Subramaniyam V, Subashchandrabose SR, Thavamani P, Megharaj M, Chen Z, Naidu R. Chlorococcum sp MM11--a novel phyco-nanofactory for the synthesis of iron nanoparticles. J Appl Phycol 2015; 27(5):1861-9. doi: 10.1007/s10811-014-0492-2 [Crossref]
  52. Bhargava A, Jain N, Barathi L M, Akhtar MS, Yun YS, Panwar J. Synthesis, characterization and mechanistic insights of mycogenic iron oxide nanoparticles. J Nanopart Res 2013; 15(11):2031. doi: 10.1007/s11051-013-2031-5 [Crossref]
  53. Bharde A, Rautaray D, Bansal V, Ahmad A, Sarkar I, Yusuf SM. Extracellular biosynthesis of magnetite using fungi. Small 2006; 2(1):135-41. doi: 10.1002/smll.200500180 [Crossref]
  54. Bharde AA, Parikh RY, Baidakova M, Jouen S, Hannoyer B, Enoki T. Bacteria-mediated precursor-dependent biosynthesis of superparamagnetic iron oxide and iron sulfide nanoparticles. Langmuir 2008; 24(11):5787-94. doi: 10.1021/la704019p [Crossref]
  55. Cornell RM, Schwertmann U. The Iron Oxides: Structure, Properties, Reactions, Occurences and Uses. 2nd ed. Weinheim: Wiley-VCH; 2003.
  56. Majewski P, Thierry B. Functionalized magnetite nanoparticles--synthesis, properties, and bio-applications. Crit Rev Solid State Mater Sci 2007; 32(3-4):203-15. doi: 10.1080/10408430701776680 [Crossref]
  57. Teja AS, Koh PY. Synthesis, properties, and applications of magnetic iron oxide nanoparticles. Prog Cryst Growth Charact Mater 2009; 55(1):22-45. doi: 10.1016/j.pcrysgrow.2008.08.003 [Crossref]
  58. Tronc E, Ezzir A, Cherkaoui R, Chanéac C, Noguès M, Kachkachi H. Surface-related properties of γ-Fe2O3 nanoparticles. J Magn Magn Mater 2000; 221(1-2):63-79. doi: 10.1016/S0304-8853(00)00369-3 [Crossref]
  59. Hendriksen PV, Linderoth S, Lindgård P. Finite-size modifications of the magnetic properties of clusters. Phys Rev B Condens Matter 1993; 48(10):7259-73. doi: 10.1103/physrevb.48.7259 [Crossref]
  60. Morales MP, Serna CJ, Bødker F, Mørup S. Spin canting due to structural disorder in maghemite. J Phys Condens Matter 1997; 9(25):5461-7. doi: 10.1088/0953-8984/9/25/013 [Crossref]
  61. Spada FE, Parker FT, Nakakura CY, Berkowitz AE. Studies of anisotropy mechanisms in polyphosphate-treated magnetic iron oxide particles. J Magn Magn Mater 1993; 120(1-3):129-35. doi: 10.1016/0304-8853(93)91304-p [Crossref]
  62. Scialabba C, Puleio R, Peddis D, Varvaro G, Calandra P, Cassata G. Folate targeted coated SPIONs as efficient tool for MRI. Nano Res 2017; 10(9):3212-27. doi: 10.1007/s12274-017-1540-4 [Crossref]
  63. Neuberger T, Schöpf B, Hofmann H, Hofmann M, von Rechenberg B. Superparamagnetic nanoparticles for biomedical applications: possibilities and limitations of a new drug delivery system. J Magn Magn Mater 2005; 293(1):483-96. doi: 10.1016/j.jmmm.2005.01.064 [Crossref]
  64. Lübbe AS, Bergemann C, Riess H, Schriever F, Reichardt P, Possinger K. Clinical experiences with magnetic drug targeting: a phase I study with 4’-epidoxorubicin in 14 patients with advanced solid tumors. Cancer Res 1996; 56(20):4686-93.
  65. Zeng J, Jing L, Hou Y, Jiao M, Qiao R, Jia Q. Anchoring group effects of surface ligands on magnetic properties of Fe₃O₄ nanoparticles: towards high performance MRI contrast agents. Adv Mater 2014; 26(17):2694-8, 016. doi: 10.1002/adma.201304744 [Crossref]
  66. Wu W, Wu Z, Yu T, Jiang C, Kim WS. Recent progress on magnetic iron oxide nanoparticles: synthesis, surface functional strategies and biomedical applications. Sci Technol Adv Mater 2015; 16(2):023501. doi: 10.1088/1468-6996/16/2/023501 [Crossref]
  67. Zhu N, Ji H, Yu P, Niu J, Farooq MU, Akram MW. Surface modification of magnetic iron oxide nanoparticles. Nanomaterials (Basel) 2018; 8(10). doi: 10.3390/nano8100810 [Crossref]
  68. Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. Biomaterials 2005; 26(18):3995-4021. doi: 10.1016/j.biomaterials.2004.10.012 [Crossref]
  69. Wu W, He Q, Jiang C. Magnetic iron oxide nanoparticles: synthesis and surface functionalization strategies. Nanoscale Res Lett 2008; 3(11):397-415. doi: 10.1007/s11671-008-9174-9 [Crossref]
  70. Couto D, Freitas M, Carvalho F, Fernandes E. Iron oxide nanoparticles: an insight into their biomedical applications. Curr Med Chem 2015; 22(15):1808-28. doi: 10.2174/0929867322666150311151403 [Crossref]
  71. Agnihotri SA, Mallikarjuna NN, Aminabhavi TM. Recent advances on chitosan-based micro- and nanoparticles in drug delivery. J Control Release 2004; 100(1):5-28. doi: 10.1016/j.jconrel.2004.08.010 [Crossref]
  72. Arakha M, Pal S, Samantarrai D, Panigrahi TK, Mallick BC, Pramanik K. Antimicrobial activity of iron oxide nanoparticle upon modulation of nanoparticle-bacteria interface. Sci Rep 2015; 5:14813. doi: 10.1038/srep14813 [Crossref]
  73. Prabaharan M. Review paper: chitosan derivatives as promising materials for controlled drug delivery. J Biomater Appl 2008; 23(1):5-36. doi: 10.1177/0885328208091562 [Crossref]
  74. Zhu A, Chan-Park MB, Dai S, Li L. The aggregation behavior of O-carboxymethylchitosan in dilute aqueous solution. Colloids Surf B Biointerfaces 2005; 43(3-4):143-9. doi: 10.1016/j.colsurfb.2005.04.009 [Crossref]
  75. Liu B, Li C, Chen G, Liu B, Deng X, Wei Y. Synthesis and optimization of MoS(2)@Fe(3)O(4)-ICG/Pt(IV) nanoflowers for MR/IR/PA bioimaging and combined PTT/PDT/chemotherapy triggered by 808 nm laser. Adv Sci (Weinh) 2017; 4(8):1600540. doi: 10.1002/advs.201600540 [Crossref]
  76. Tong S, Quinto CA, Zhang L, Mohindra P, Bao G. Size-dependent heating of magnetic iron oxide nanoparticles. ACS Nano 2017; 11(7):6808-16. doi: 10.1021/acsnano.7b01762 [Crossref]
  77. Darini A, Eslaminejad T, Nematollahi Mahani SN, Ansari M. Magnetogel nanospheres composed of cisplatin-loaded alginate/B-cyclodextrin as controlled release drug delivery. Adv Pharm Bull 2019; 9(4):571-7. doi: 10.15171/apb.2019.065 [Crossref]
  78. Mumtaz S, Wang LS, Hussain SZ, Abdullah M, Huma Z, Iqbal Z. Dopamine coated Fe(3)O(4) nanoparticles as enzyme mimics for the sensitive detection of bacteria. Chem Commun (Camb) 2017; 53(91):12306-8. doi: 10.1039/c7cc07149c [Crossref]
  79. Rajkumar S, Prabaharan M. Multi-functional nanocarriers based on iron oxide nanoparticles conjugated with doxorubicin, poly(ethylene glycol) and folic acid as theranostics for cancer therapy. Colloids Surf B Biointerfaces 2018; 170:529-37. doi: 10.1016/j.colsurfb.2018.06.051 [Crossref]
  80. Yee C, Kataby G, Ulman A, Prozorov T, White H, King A. Self-assembled monolayers of alkanesulfonic and -phosphonic acids on amorphous iron oxide nanoparticles. Langmuir 1999; 15(21):7111-5. doi: 10.1021/la990663y [Crossref]
  81. Sahoo Y, Pizem H, Fried T, Golodnitsky D, Burstein L, Sukenik CN. Alkyl phosphonate/phosphate coating on magnetite nanoparticles: a comparison with fatty acids. Langmuir 2001; 17(25):7907-11. doi: 10.1021/la010703+ [Crossref]
  82. Soares PI, Lochte F, Echeverria C, Pereira LC, Coutinho JT, Ferreira IM. Thermal and magnetic properties of iron oxide colloids: influence of surfactants. Nanotechnology 2015; 26(42):425704. doi: 10.1088/0957-4484/26/42/425704 [Crossref]
  83. Costa C, Brandão F, Bessa MJ, Costa S, Valdiglesias V, Kiliç G. In vitro cytotoxicity of superparamagnetic iron oxide nanoparticles on neuronal and glial cells Evaluation of nanoparticle interference with viability tests. J Appl Toxicol 2016; 36(3):361-72. doi: 10.1002/jat.3213 [Crossref]
  84. Toropova YG, Golovkin AS, Malashicheva AB, Korolev DV, Gorshkov AN, Gareev KG. In vitro toxicity of Fe(m)O(n), Fe(m)O(n)-SiO(2) composite, and SiO(2)-Fe(m)O(n) core-shell magnetic nanoparticles. Int J Nanomedicine 2017; 12:593-603. doi: 10.2147/ijn.s122580 [Crossref]
  85. Malvindi MA, De Matteis V, Galeone A, Brunetti V, Anyfantis GC, Athanassiou A. Toxicity assessment of silica coated iron oxide nanoparticles and biocompatibility improvement by surface engineering. PLoS One 2014; 9(1):e85835. doi: 10.1371/journal.pone.0085835 [Crossref]
  86. Ivashchenko O, Lewandowski M, Peplińska B, Jarek M, Nowaczyk G, Wiesner M. Synthesis and characterization of magnetite/silver/antibiotic nanocomposites for targeted antimicrobial therapy. Mater Sci Eng C Mater Biol Appl 2015; 55:343-59. doi: 10.1016/j.msec.2015.05.023 [Crossref]
  87. Bisht G, Rayamajhi S, Kc B, Paudel SN, Karna D, Shrestha BG. Synthesis, characterization, and study of in vitro cytotoxicity of ZnO-Fe3O4 magnetic composite nanoparticles in human breast cancer cell line (MDA-MB-231) and mouse fibroblast (NIH 3T3). Nanoscale Res Lett 2016; 11(1):537. doi: 10.1186/s11671-016-1734-9 [Crossref]
  88. Sun Q, Li J, Le T. Zinc oxide nanoparticle as a novel class of antifungal agents: current advances and future perspectives. J Agric Food Chem 2018; 66(43):11209-20. doi: 10.1021/acs.jafc.8b03210 [Crossref]
  89. Lemire JA, Harrison JJ, Turner RJ. Antimicrobial activity of metals: mechanisms, molecular targets and applications. Nat Rev Microbiol 2013; 11(6):371-84. doi: 10.1038/nrmicro3028 [Crossref]
  90. Palza H. Antimicrobial polymers with metal nanoparticles. Int J Mol Sci 2015; 16(1):2099-116. doi: 10.3390/ijms16012099 [Crossref]
  91. Zhang YM, Rock CO. Membrane lipid homeostasis in bacteria. Nat Rev Microbiol 2008; 6(3):222-33. doi: 10.1038/nrmicro1839 [Crossref]
  92. Studer AM, Limbach LK, Van Duc L, Krumeich F, Athanassiou EK, Gerber LC. Nanoparticle cytotoxicity depends on intracellular solubility: comparison of stabilized copper metal and degradable copper oxide nanoparticles. Toxicol Lett 2010; 197(3):169-74. doi: 10.1016/j.toxlet.2010.05.012 [Crossref]
  93. Behera SS, Patra JK, Pramanik K, Panda N, Thatoi H. Characterization and evaluation of antibacterial activities of chemically synthesized iron oxide nanoparticles. World J Nano Sci Eng 2012; 2(4):196-200. doi: 10.4236/wjnse.2012.24026 [Crossref]
  94. 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; 79(1):113-6. doi: 10.1016/j.chemosphere.2009.12.056 [Crossref]
  95. Saleh NB, Chambers B, Aich N, Plazas-Tuttle J, Phung-Ngoc HN, Kirisits MJ. Mechanistic lessons learned from studies of planktonic bacteria with metallic nanomaterials: implications for interactions between nanomaterials and biofilm bacteria. Front Microbiol 2015; 6:677. doi: 10.3389/fmicb.2015.00677 [Crossref]
  96. Pelgrift RY, Friedman AJ. Nanotechnology as a therapeutic tool to combat microbial resistance. Adv Drug Deliv Rev 2013; 65(13-14):1803-15. doi: 10.1016/j.addr.2013.07.011 [Crossref]
  97. Sharaf SMA, Abbas HS, Ismaeil TAM. Characterization of spirugenic iron oxide nanoparticles and their antibacterial activity against multidrug-resistant Helicobacter pylori. Egypt J Phycol 2019; 20:1-28.
  98. Leuba KD, Durmus NG, Taylor EN, Webster TJ. Short communication: Carboxylate functionalized superparamagnetic iron oxide nanoparticles (SPION) for the reduction of S aureus growth post biofilm formation. Int J Nanomedicine 2013; 8:731-6. doi: 10.2147/ijn.s38256 [Crossref]
  99. Niemirowicz K, Swiecicka I, Wilczewska AZ, Misztalewska I, Kalska-Szostko B, Bienias K. Gold-functionalized magnetic nanoparticles restrict growth of Pseudomonas aeruginosa. Int J Nanomedicine 2014; 9:2217-24. doi: 10.2147/ijn.s56588 [Crossref]
  100. Reddy PM, Chang KC, Liu ZJ, Chen CT, Ho YP. Functionalized magnetic iron oxide (Fe3O4) nanoparticles for capturing gram-positive and gram-negative bacteria. J Biomed Nanotechnol 2014; 10(8):1429-39. doi: 10.1166/jbn.2014.1848 [Crossref]
  101. Palza H, Gutiérrez S, Delgado K, Salazar O, Fuenzalida V, Avila JI. Toward tailor-made biocide materials based on poly(propylene)/copper nanoparticles. Macromol Rapid Commun 2010; 31(6):563-7. doi: 10.1002/marc.200900791 [Crossref]
  102. Thukkaram M, Sitaram S, Kannaiyan SK, Subbiahdoss G. Antibacterial efficacy of iron-oxide nanoparticles against biofilms on different biomaterial surfaces. Int J Biomater 2014; 2014:716080. doi: 10.1155/2014/716080 [Crossref]
  103. Maleki Dizaj S, Lotfipour F, Barzegar-Jalali M, Zarrintan MH, Adibkia K. Antimicrobial activity of the metals and metal oxide nanoparticles. Mater Sci Eng C Mater Biol Appl 2014; 44:278-84. doi: 10.1016/j.msec.2014.08.031 [Crossref]
  104. Hussein-Al-Ali SH, El Zowalaty ME, Kura AU, Geilich B, Fakurazi S, Webster TJ. Antimicrobial and controlled release studies of a novel nystatin conjugated iron oxide nanocomposite. Biomed Res Int 2014; 2014:651831. doi: 10.1155/2014/651831 [Crossref]
  105. Niemirowicz K, Durnaś B, Tokajuk G, Głuszek K, Wilczewska AZ, Misztalewska I. Magnetic nanoparticles as a drug delivery system that enhance fungicidal activity of polyene antibiotics. Nanomedicine 2016; 12(8):2395-404. doi: 10.1016/j.nano.2016.07.006 [Crossref]
  106. Prucek R, Tuček J, Kilianová M, Panáček A, Kvítek L, Filip J. The targeted antibacterial and antifungal properties of magnetic nanocomposite of iron oxide and silver nanoparticles. Biomaterials 2011; 32(21):4704-13. doi: 10.1016/j.biomaterials.2011.03.039 [Crossref]
  107. Wilczewska AZ, Kosińska A, Misztalewska-Turkowicz I, Kubicka A, Niemirowicz-Laskowska K, Markiewicz KH. Magnetic nanoparticles bearing metallocarbonyl moiety as antibacterial and antifungal agents. Appl Surf Sci 2019; 487:601-9. doi: 10.1016/j.apsusc.2019.05.159 [Crossref]
Submitted: 21 Oct 2019
Revised: 06 Feb 2020
Accepted: 10 Feb 2020
First published online: 08 Aug 2020
EndNote EndNote

(Enw Format - Win & Mac)

BibTeX BibTeX

(Bib Format - Win & Mac)

Bookends Bookends

(Ris Format - Mac only)

EasyBib EasyBib

(Ris Format - Win & Mac)

Medlars Medlars

(Txt Format - Win & Mac)

Mendeley Web Mendeley Web
Mendeley Mendeley

(Ris Format - Win & Mac)

Papers Papers

(Ris Format - Win & Mac)

ProCite ProCite

(Ris Format - Win & Mac)

Reference Manager Reference Manager

(Ris Format - Win only)

Refworks Refworks

(Refworks Format - Win & Mac)

Zotero Zotero

(Ris Format - FireFox Plugin)

Abstract View: 1153
PDF Download: 738
Full Text View: 254

© 2011-2024 Tabriz University of Medical Sciences; unless otherwise stated. This Platinum Open Access journal is a title of TUOMS PRESS; an imprint of Tabriz University of Medical Sciences.

Journal Management System. Powered by Maad Rayan