year 15, Issue 1 (January - February 2021)                   Iran J Med Microbiol 2021, 15(1): 18-45 | Back to browse issues page


XML Persian Abstract Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Ghaderi R S, Kazemi M, Soleimanpour S. Nanoparticles are More Successful Competitor than Antibiotics in Treating Bacterial Infections: A Review of the Literature. Iran J Med Microbiol 2021; 15 (1) :18-45
URL: http://ijmm.ir/article-1-1125-en.html
1- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
2- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
3- Chemistry Department, Payam-e Noor University, Mashhad, Iran , mo.kazemi2009@yahoo.com
Full-Text [PDF 1793 kb]   (2804 Downloads)     |   Abstract (HTML)  (4444 Views)
Full-Text:   (1634 Views)
Introduction

.

The emergence of antibiotic resistance in bacteria is one of the most important global health issues, and new research is needed around the world to develop more effective antimicrobial compounds (1). In the 21st century, nanotechnology has become one of the most important and influential technologies in science worldwide. Nanomaterial engineering can open up new avenues of research and development in some fields including medicine, cosmetics, agriculture, environmental sciences, materials science, biomedical sciences, information, and food technology. It is also used to understand the changes in the physicochemical properties of nanomaterials (5). In fact, nanotechnology covers a wide range of sciences. In developing countries, it is important to invest and focus on this science to make improvements in the fields of health, water, energy, and environment (6,7). Nanomaterials are structures that have at least one dimension on the nanometer scale (1-100 nanometers). On a broad spectrum, nanoparticles have shown impacts against both gram-positive and gram-negative bacteria (8). For example, zinc oxide nanoparticles could inhibit Staphylococcus aureus and silver nanoparticles have shown concentration-dependent antimicrobial activity against E. coli and Pseudomonas aeruginosa (9). Recently, nanoparticles (NPs) have drawn much attention because of their potential to be an alternative to antibiotics. The unique properties of nanoparticles compared to the balk form make them ideal for diagnosis, treatment, and antibiotic delivery systems. Rapid and sensitive detection of bacteria can be done by nanoparticle-based methods. In nanotechnology, there are new perspectives for the development of new formulations based on different types of nanoparticles with different sizes, shapes, and antimicrobial properties (11). Nanoparticles might seem to be a promising solution because not only can they fight against bacteria, but they can also act as carriers of antibiotics and natural antimicrobial compounds. This study focuses on the mechanisms of bacterial resistance and antibacterial activity of nanoparticles because it is very important to investigate the antibacterial mechanisms of these particles to produce more effective antimicrobial agents.

 
 

Materials and Methods

Standard requirements for literature reviews were conducted in the English language listed on health and medical electronic databases. The search words were: "Bacteria,” “Nanotechnology,” “Antibacterial,” and “Nano” using PubMed, Scopus, Science Direct, and Google Scholar databases. Furthermore, manual searches of other relevant journals and keywords searches were performed. We have focused on published papers from 2010 to2020. After the initial review, the following types of articles were excluded: (i) studies that were not relevant to the main subject, (ii) papers which their full texts were not available, and (iii) repetitive publications and articles with similar results. We included articles that discussed the following topics: Articles that have examined the antibacterial mechanisms of nanoparticles on bacteria, as well as articles that explored the benefits and application of nanoparticles to combat bacteria, and finally studies that examined factors that affect the antibacterial mechanisms of nanoparticles.


 

Results

Mechanisms of Bacterial Resistance to Antibiotics
Bacterial resistance occurs due to the widespread use of antibiotics in prevention or treatment of diseases without proper medical symptoms, which has become a serious problem. This resistance has several reasons, all of which occurred due to the interaction of internal and external factors (12). External factors usually include antibiotics pressure and human-caused environmental changes, and internal factors include resistance from the genetic point of view and at the DNA level, as well as resistance from the biochemical point of view and at the protein level (14,15).

Advantages of using Nanoparticles in treating Bacterial Infections
Bacteria can develop resistance to antimicrobials. Nanoparticles can combat microbes through various mechanisms that are simultaneously activated. Simultaneous mechanisms greatly reduce the possibility of several mutations in different genes, and most antibiotic resistance mechanisms are irrelevant to nanoparticle resistance mechanism; therefore, it is hoped that bacteria will be less resistant to nanoparticles (17). Nanoparticles are widely affected by the immune system. Many studies have shown that they have the ability to strengthen the immune system. The produced nanoparticles enter the body as a foreign substance, in return, the innate immune system responds immediately to these substances. This response can vary depending on the physicochemical properties of nanoparticles, including the size, charge, shape, and degree of hydrophobicity. As a result, the interaction of nanoparticles with various components of the immune system such as neutrophils, soluble proteins, and antigen-processing cells (APCs) depends on nanoparticle properties (18). Another case of stimulation of the immune system by nanoparticles is the development of allergic reactions. There are also many immune system reactions triggered by nanoparticles, which are produced by the production of inflammatory cytokines. Numerous studies have reported the induction of cytokines with different types of nanomaterials (gold colloids, dendrimers, polymers, lipid nanoparticles, etc.). Based on the body's immune response to nanoparticles, these substances can be used to treat cancers and autoimmune diseases, and act as a vaccine (19-20). Information about the different effects of nanoparticles on cells of the innate immune system (macrophages, dendritic cells, neutrophils, mast cells, and natural killer cells) and cells of the adaptive immune system (T cells and B cells) helps us understand the immunological effects of nanomaterials. Innate immune system cells such as macrophages and dendritic cells cause the expression of inflammatory cytokines, activation of T cell lymphocytes and activation of inflammasome. In addition, Nanoparticles can secrete histamine and increase cytosolic calcium ions by affecting on mast cells. Also nanoparticles are able to target natural killer cells (NK cell) to control their movement toward the tumor. By affecting the B cells of the adaptive immune system, the nanoparticles enhance immunity during vaccination and help target B-cell lymphoma without the need for chemotherapy. The interaction of nanoparticles with T lymphocytes can lead to Th1 and Th2 responses as well as cytokine production and it helps development of nanoparticle-based products. Moreover, nanoparticles can act as a carrier for antibiotics synergistic factor and as a transvestite factor, which can ultimately facilitate therapeutic efficacy (21,22). The combination of nanoparticles (NPs) and antimicrobial materials, such as antibiotics, peptides, or different biomolecules, has been known as one of the most effective methods targeting antibiotic resistance. Due to their small size, they can be used to treat infections caused by intracellular pathogens (24,25). They also protect the drugs from the effects of enzymes and harmful chemical reactions. Nanoparticle carriers can help accurately target the infection site through antibiotics. Therefore, the unwanted side effects due to high doses and systemic side effects are minimized. With the use of these types of carriers, the drug concentration at the site of infection can reach the required effective level and be stable for a long time. Carrier nanoparticles can combine and transmit several antibacterial drugs together (26-29).

The Problems of using Nanoparticles
Bacterial Resistance to Nanoparticles
Further research has identified new aspects regarding the effects of nanoparticles on bacteria. While most previous studies have mentioned that nanoparticles, as an antibacterial agent, prevent the development of resistant bacteria, new information has recently been found to support the opposite. One study reported the increase in plasmid transport (such as RP4, PK2, and pCF10) by aluminum nanoparticles, which leads to the formation of resistant strains (30).

Toxicity
Local and systemic toxicity, as well as harmful effects on beneficial bacteria in the human body, are among the concerns discussed regarding the use of nanoparticles. Some NPs can cause hemolysis and interfere with blood coagulation pathways. One study has shown the deposition of silver nanoparticles in the liver, spleen, lungs, and other organs, which leads to organ damage. Intravenous nanoparticles may accumulate in the large intestine, lungs, bone marrow, liver, spleen, and lymphatic system, and their inhalation can cause toxicity in the lungs. Besides, several studies showed that nanoparticles do not have any dangerous toxicity in vivo conditions. The exact mechanism of the toxic effects of nanoparticles in laboratory conditions is unclear; therefore, further studies are required (31).

Application of Nanoparticles
Nanoparticles are used in different fields due to their unique properties and their effect on the bacteria. Different Types of nanoparticles have their characteristics, and each type is used for antibacterial purposes in certain parts of the body. Currently, there are some undergoing clinical trials on drugs with metal-based nanoparticles against bacterial infections with name Arikace, Lipoquin, Pulmaquin, Silvasorb, MAT2501 and QA-PEI. The cost of producing and using these nanoparticles is very high compared to traditional medicines; therefore, conventional treatments are more commonly used. Finally, nanoparticle-based drugs may be preferred in specific conditions and can increase the patients' quality of life (32).

Antibacterial Mechanisms of Nanoparticles
Despite special attention to nanomaterials, their antibacterial mechanisms have not been well known. But the accepted and important antibacterial mechanisms of nanoparticles against bacteria will be discussed here.

The Function of Nanoparticles due to Oxidative Stress
Oxidative stress induced by ROS is an important antibacterial mechanism of NPs. It can attack proteins and reduce the activity of some periplasmic enzymes. Moreover, reactive oxygen species are effective in increasing gene expression levels of oxidative proteins which are considered an important mechanism in bacterial cell apoptosis (33-34). It can destroy cellular components of the pathogens like membrane, DNA, and mitochondria, which can result in cell death (35). The study carried out by Ansari et al. confirmed that Al2O3 nanoparticles pass through the cell membrane and enter the cell, then nanoparticle-membrane interaction and due to intracellular oxidative stress, membrane integrity is damaged (36). Nanoparticles can also bind to TrxR and Trx active sites and interfere with the thioredoxin system, which is one of the most important redox systems of disulfide that is used by bacteria against oxidative stress, and leads to oligomerization and dysfunction of TrxR and TrxR (37).

Performance of Nanoparticles by Producing Soluble Metal Ions
The next important mechanism is releasing metal ions from metal oxide. Metal ions are slowly released from metal oxide and then absorbed through the cell membrane, followed by direct interaction with functional groups such as proteins and nucleic acids. This interaction alters cell structure, disrupts enzymatic activity, and interrupts the common physiological activity in bacterial cells (42).

Mechanisms of Nanoparticles Penetration into Bacteria
Diffusion: Nanoparticles can transmit reactive oxygen species to bacteria using diffusion. In a study, Pan et al. showed that iron oxide-graphene oxide nanoparticles could inactivate methicillin-resistant Staphylococcus aureus (MRSA) due to the production of large amounts of hydroxyl radicals and diffusion into bacterial cells (43).
Adsorption: Metal nanoparticles are released into the environment and bind to negatively charged groups in the bacterial cell membrane, such as carboxylic and phosphate groups, in a process known as biological adsorption. In a study, it was shown that the surface charges of copper nanoparticles significantly affect the adsorption of nanoparticles to membranes (46).

 

Figure 1. The nanoparticles application in medicine
Figure 1. The nanoparticles application in medicine
 

The Effect of Nanoparticles on Different Parts of Bacteria
Interaction of Nanoparticles with the Cell Membrane and Bacterial Cell Wall
Studies showed that the interaction of nanoparticles with cell membranes in gram-positive and gram-negative bacteria is different due to their specific structure. Lipopolysaccharide (LPS) is a unique structure of the cell wall of gram-negative bacteria that creates a negatively charged region and absorbs nanoparticles. In contrast, teichoic acid is only expressed in the cell wall of gram-positive bacteria; therefore, nanoparticles are distributed along with the phosphate molecular structure and prevent their accumulation. Many studies have shown that nanoparticles have more antibacterial activity against gram-positive bacteria in comparison with gram-negative bacteria because the cell wall of gram-negative bacteria is composed of lipopolysaccharides, lipoproteins, and phospholipids. Nanoparticles can transmit reactive species of oxygen into bacteria through diffusion (47). The results of a study by Lucanhan et al. showed that selenium nanoparticles have a strong electrostatic repulsion compared to lipopolysaccharide and gram-negative bacterial membranes due to their very negative nature. Gram-positive bacteria, on the other hand, have significantly less negative charge than gram-negative bacteria. Therefore, selenium nanoparticles are more likely to be deposited on the surface of gram-positive bacteria and cause bacterial death (39). Foster et al. confirmed that titanium oxide nanoparticles can adhere to the surface of bacterial cells to produce reactive oxygen species and damage the composition and structure of cell membranes, thereby they disrupt cell membrane function and cause cell leakage, and ultimately kill the bacteria (50). One of the most important functions of the cell membrane is the respiratory activity of bacteria. Studies have reported that nanoparticles also disrupt the respiratory activity of bacterial cell membranes (52). Figure 2 shows the attachment, penetration and cell lysis of bacteria with Kan-AuNPs treatment (83)
 
 
 Figure 2. morphological changes in bacteria upon treating with Kan-AuNPs at different intervals of time. A) S. epidermidis bacteria treated with Kan-AuNPs B) E. aerogenes bacteria treated with Kan-AuNPs, Kan-AuNPs were found to attach and penetrate to the bacterial cell wall, disrupting the cellular environment and leading to cell lysis due to leakage of cellular components (83).
Figure 2. morphological changes in bacteria upon treating with Kan-AuNPs at different intervals of time. A) S. epidermidis bacteria treated with Kan-AuNPs B) E. aerogenes bacteria treated with Kan-AuNPs, Kan-AuNPs were found to attach and penetrate to the bacterial cell wall, disrupting the cellular environment and leading to cell lysis due to leakage of cellular components (83).
 
 
Inhibition of Protein and DNA Synthesis in Bacteria
 In recent years, the effect of nanoparticles in the synthesis of bacterial proteins has received much attention from researchers. Su et al. investigated the effect of copper nanoparticles on nitrification of bacteria by maQing a significant change in the expression of key proteins. After entering the cell, proteomic bioinformatics analysis showed that copper nanoparticles regulate proteins involved in nitrogen metabolism, electron transfer, and material transport (54). Iron oxide nanoparticles were studied based on their strong tendency to form disulfide bonds and it was shown that they affect the metabolism and redox status of bacterial cells.
Nanoparticles can also stop protein and DNA synthesis in bacteria by inhibiting the combination of ribosomal subunits with tRNA or restricting ATPase activity to reduce ATP levels (55-56). Studies have indicated that nanoparticles can bind to proteins in cell membranes and create stable bonds. And as a result, they inactivate proteins that are involved in the production of membrane ATP and mediate the transfer of ions through cell membranes (10).

Regulation of the Expression of Metabolic Genes by Nanoparticles
The expression of metabolic genes is regulated by nanoparticles. Nanoparticles can also regulate cell membrane penetration and interfere with molecular pathways. Bacterial metabolic processes play a major role in bacterial growth and reproduction. They can also cause pathogenicity in bacteria. Therefore, targeted alteration of bacterial metabolic activity can be used to regulate bacterial pathogenicity For example, it has been observed that copper oxide nanoparticles can regulate the expression of proteins involved in bacterial nitrogen metabolism and significantly inhibit the activity of nitrate reductase and nitrite reductase. Wang et al. reported that silver nanoparticles can be covalently bonded to thiol derivatives and attached to some genetic components without any complicated modification process (47-48).

Inhibition of Bacterial Biofilms Formation by Nanoparticles
Prevention of biofilm formation by nanoparticles is an important mechanism because biofilms play an important role in the development of bacterial resistance. Genotypic and phenotypic properties of cells in biofilms are different from those of free cells, and these differences make them highly resistant to antibiotics (35). By the use of silver nitrate (AgNO3), Mohanti was able to produce silver nanoparticles with proper anti-biofilm effects (60). Pan et al. also reported that nanoparticles could affect the metabolic rate of bacterial communities. Bacterial metabolism is considered an important activity for biofilms. Based on other studies, nanoparticles can prevent the formation of biofilm by preventing bonding and icaAD transcription. Smaller nanoparticles are more capable of destroying bacterial biofilms due to the increase in surface-to-mass ratio (61). Due to the small size of nanoparticles, they can penetrate microbial cell walls and biofilm layers and cause irreversible damages to cell membranes and DNA. The shape of nanoparticles affects the destruction of biofilms. for instance, rod-like nanoparticles are more effective at destroying biofilms than spherical nanoparticles (64).
 
 
 Figure 3. Antibacterial mechanisms of nanoparticles
Figure 3. Antibacterial mechanisms of nanoparticles
 
Table 1. The antibacterial mechanisms of some nanoparticles against pathogenic bacteria. (46,76–82).
Mechanisms of antibacterial action Targeted bacteria nanoparticles
-Interference with electron and energy transfer through the membrane
-Prevention of DNA replication and electron transport chain in bacteria and fungi
-Disruption of the cell surface and loss of membrane integrity
-Penetration into bacterial biofilms using an external magnetic field
-Generation of reactive oxygen species (ROS)
-Combination with antibiotics such as vancomycin, tetracycline and ampicillin
Methicillin-resistant Staphylococcus aureus, Staphylococcus epidermidis. Vancomycin-resistant Enterococcus faecium and Klebsiella pneumoniae Ag Nps
-Generation of reactive oxygen species (ROS), lipid peroxidation, alkaline, electrostatic interference S. aureus, E. coli, Bacillus megaterium, Bacillus subtilis MgO NPs
-Generation of reactive oxygen species (ROS), superoxide radicals
-release of ion and binding to the thiol group in bacterial surface proteins
-DNA damage
S. aureus, E. coli TiO2 NPs
-Generation of reactive oxygen species (ROS)
-Penetration of hydrogen peroxide produced at the surface of nanoparticles into bacterial cells
-Prevention of enzymes product
-production of Zn2+, disruption of membrane and interference with intracellular components
S. aureus, E. coli, listeria monocytogenes, Salmonela Zn NPs
-Generation of reactive oxygen species (ROS)
-Creating a hole in the bacterial cell wall
-DNA binding, DNA degradation and prevent transcription steps
-Osmotic balance and integrity of the bacterial cell wall
-Penetration into the biofilm
-Combined with antibiotics
Methicillin-resistant S. aureus (MRSA), Proteus mirabilis, A. baumannii ,E. coli, P. aeruginosa, S. aureus Au NPs
-Generation of oxidative stress through the production of reactive oxygen species (ROS) -superoxide radical -hydroxyl radical -hydrogen peroxide -oxygen S. aureus, E. coli, S. epidermidis Fe NPs
-Generation of reactive oxygen species (ROS)
-Cell wall disruption
E. coli Al NPs
-Changing in Krebs cycle
-Interference with the metabolism of amino acids and nucleotides
Helicobacter pylori Bi NPs
-Prevent energy metabolism
-Interference with electron transport chain

-Cell wall disruption
E.coli, Salmonella enteric, E. faecium, Streptococcus spp., Shewanella oneidensis, Acinetobacter baumannii, Burkholderia cepacia, Yersinia pestis, and K. pneumonia carbon-based NPs
-Generation of reactive oxygen species (ROS)
-Interference with biochemical processes within bacterial cells
S. aureus, E. coli, B. subtilis CuO NPs
-Generation of reactive oxygen species (ROS)
- Prevent the growth of bacteria
E. coli, P.aeruginosa, S. aureus Se NPs


Factors affecting the Antibacterial Mechanisms of Nanoparticles
Physicochemical properties of nanoparticles including size, charge, zeta potential, surface morphology, and crystal structure, are important factors regulating the performance of nanoparticles on bacterial cells. Also, environmental conditions and duration of exposure to other factors affect the antibacterial effects of nanoparticles. (65). Current research has shown that the size of a nanoparticle greatly affects its antibacterial activity. Smaller nanoparticles have more specific surface areas and are more likely to come in contact with bacterial cell membranes compared to large nanoparticles and polymers (66). Since the shape is an important factor in antimicrobial activity, nanoparticles with different shapes can damage bacterial cells by interacting with pre-plasmic enzymes. Although nanoparticles are reported to have different forms such as bars, wires, tubes, scales, and discs, they are mostly seen spherically. Nanotubes and nanorods seem to be more effective than other forms to kill the bacteria. This indicates that the effect of antibacterial activity is due to the specific surface area (67-68). By increasing the roughness of nanoparticles, the size and ratio of surface to mass causes the absorption of bacterial proteins and consequently reduces the bacterial adhesion of nanoparticles (69). Recent studies have shown that surface charge in nanoparticles has a significant impact on the association between bacteria and nanoparticles. This connection is due to the electrostatic attraction between positively charged nanoparticles and negatively charged bacterial cell membranes. However, at higher concentrations, negatively charged nanoparticles have a good level of antibacterial activity due to molecular overcrowding, which leads to proper interaction between nanoparticles and bacterial cells (70). For example, positively charged polystyrene nanoparticles disrupt the electron transfer chain in bacteria. A study on E. coli bacteria also reported that bacteria with mutations in the ubiquinone gene were more sensitive to positively charged nanoparticles (67). A wide range of studies has also shown that different environmental conditions cause significant differences in the antimicrobial activity of nanoparticles. For instance, ambient temperature has different impacts on antibacterial activity due to its effects on the production of reactive oxygen species. Additionally, the pH of the environment affects antimicrobial activity. It appears that acidic conditions bind nanoparticles to the bacterial cell walls. Furthermore, for example with decreasing pH, the rate of dissolution of zinc oxide nanoparticles increases, which causes an increase in antimicrobial properties (73). According to studies, nanoparticles are a good alternative to antibiotics and have a high potential to solve the problem of the emergence of resistant bacteria; because some nanoparticles have no cellular toxicity or they have very low toxicity. Also, new methods for nanoparticle production do not involve high-risk and complex processes (74).

Research Limits
The antibacterial mechanisms of nanoparticles are still unknown. Many studies, for example, attribute antibacterial activity to oxidative stress or reactive oxygen species, while for some nanoparticles, including magnesium oxide nanoparticles, the antibacterial mechanism may not regulate bacterial metabolism; therefore, one of the issues to be addressed in future studies is to further investigate the antibacterial mechanisms of nanoparticles. Lack of integrated standards is another limitation regarding antibacterial mechanisms of nanoparticles. In particular, the use of different bacterial strains, operating time, and nanoparticle properties have not been standardized in various studies, which makes it difficult to compare antibacterial activities. In addition, sensitive bacterial strains are often used to accurately determine the antibacterial activity of nanoparticles. Other limitations include the complex structure of bacterial cell membranes and the lack of research methods for their laboratory studies. Moreover, laboratory models cannot replicate in vivo conditions accurately; therefore, it is impossible to estimate the antibacterial performance of nanoparticles through laboratory bacterial cell culture alone. Many questions remain unanswered, questions regarding the toxicity of nanoparticles as well as how nanoparticles pass through the bacterial cell membrane which all will need to be answered in future researches.

 
 
Conclusion

Since in recent years antibiotic resistance has accelerated, fighting infectious diseases and treating patients have become challenging, which leads to serious complications and death. According to several studies, nanoparticles are a suitable replacement for antibiotics and it seems they could solve the problem of the emergence of resistant bacteria; since nanoparticles either have no cytotoxicity or are usually very low in toxicity. Also, their production methods do not involve high-risk and complex processes. Nowadays, with the help of green chemistry, smaller and more efficient nanoparticles are produced by simple and clean methods with acceptable antibacterial properties (71).
We hope that this review study can provide a better perspective for researchers and an in-depth study of the antibacterial mechanisms of nanoparticles will lead to the production of effective antibacterial particles with no cytotoxicity.


 

Acknowledgements

The technical support for this work has been provided by Mashhad University of Medical Sciences.

 
 

Compliance with Ethical Standards

The authors declare that they have no conflict of interest.

 

Conflicts of Interest

Authors declared no conflict of interests.


 

Type of Study: Review Article | Subject: Antimicrobial Substances
Received: 2020/05/3 | Accepted: 2020/11/10 | ePublished: 2021/01/10

References
1. Ghaderi R, Yaghoubi A, Hashemy I, Ghazvini K. The prevalence of genes encoding ESBL among clinical isolates of Escherichia coli in Iran: A systematic review and meta-analysis. Gene Reports. 2019;100562. [DOI:10.1016/j.genrep.2019.100562]
2. Hsueh P-R. New Delhi metallo-ss-lactamase-1 (NDM-1): an emerging threat among Enterobacteriaceae. Vol. 109, Journal of the Formosan Medical Association = Taiwan yi zhi. Singapore; 2010. p. 685-7. [DOI:10.1016/S0929-6646(10)60111-8]
3. Poole K. Mechanisms of bacterial biocide and antibiotic resistance. 2002;(Levy 2000):55-64. [DOI:10.1046/j.1365-2672.92.5s1.8.x]
4. Knetsch MLW, Koole LH. New Strategies in the Development of Antimicrobial Coatings: The Example of Increasing Usage of Silver and Silver Nanoparticles. 2011;340-66. [DOI:10.3390/polym3010340]
5. Gunti L, Dass RS, Kalagatur NK. Phytofabrication of Selenium Nanoparticles From Emblica officinalis Fruit Extract and Exploring Its Biopotential Applications : Antioxidant , Antimicrobial , and Biocompatibility. 2019;10(April):1-17. [DOI:10.3389/fmicb.2019.00931] [PMID] [PMCID]
6. Journal AI, Sowndarya P, Ramkumar G, Shivakumar MS. Green synthesis of selenium nanoparticles conjugated Clausena dentata plant leaf extract and their insecticidal potential against mosquito vectors. Artif Cells, Nanomedicine, Biotechnol [Internet]. 2017;0(0):1490-5. Available from: [DOI:10.1080/21691401.2016.1252383] [PMID]
7. Han J, Zhao D, Li D, Wang X, Jin Z, Zhao K. Polymer-based nanomaterials and applications for vaccines and drugs. Polymers (Basel). 2018;10(1):31. [DOI:10.3390/polym10010031] [PMID] [PMCID]
8. Kazemi M. Evaluation of Antifungal and Photocatalytic Activities of Gelatin-Stabilized Evaluation of Antifungal and Photocatalytic Activities of Gelatin ‑ Stabilized Selenium Oxide Nanoparticles. J Inorg Organomet Polym Mater [Internet]. 2020;(February). Available from: [DOI:10.1007/s10904-020-01462-4]
9. Ramalingam B, Parandhaman T, Das SK. Antibacterial Effects of Biosynthesized Silver Nanoparticles on Surface Ultrastructure and Nanomechanical Properties of Gram-Negative Bacteria viz. Escherichia coli and Pseudomonas aeruginosa. ACS Appl Mater Interfaces. 2016 Feb;8(7):4963-76. [DOI:10.1021/acsami.6b00161] [PMID]
10. Nagy A, Harrison A, Sabbani S, Munson RSJ, Dutta PK, Waldman WJ. Silver nanoparticles embedded in zeolite membranes: release of silver ions and mechanism of antibacterial action. Int J Nanomedicine. 2011;6:1833-52. [DOI:10.2147/IJN.S24019] [PMID] [PMCID]
11. Kolhatkar AG, Jamison AC, Litvinov D, Willson RC, Lee TR. Tuning the Magnetic Properties of Nanoparticles. 2013. [DOI:10.1002/chin.201451225]
12. Aung MS, Zi H, Nwe KM, Maw WW, Aung MT, Min WW, et al. Drug resistance and genetic characteristics of clinical isolates of staphylococci in Myanmar: high prevalence of PVL among methicillin-susceptible Staphylococcus aureus belonging to various sequence types. New microbes new Infect. 2016 Mar;10:58-65. [DOI:10.1016/j.nmni.2015.12.007] [PMID] [PMCID]
13. Mehdipour Moghaddam MJ, Mirbagheri AA, Salehi Z, Habibzade SM. Prevalence of Class 1 Integrons and Extended Spectrum Beta Lactamases among Multi-Drug Resistant Escherichia coli Isolates from North of Iran. Iran Biomed J. 2015;19(4):233-9.
14. Kazemi M, Akbari A, Zarrinfar H, Soleimanpour S, Sabouri Z, Khatami M, et al. Evaluation of Antifungal and Photocatalytic Activities of Gelatin-Stabilized Selenium Oxide Nanoparticles. J Inorg Organomet Polym Mater [Internet]. 2020; Available from: [DOI:10.1007/s10904-020-01462-4]
15. Khameneh B, Diab R, Ghazvini K, Fazly Bazzaz BS. Breakthroughs in bacterial resistance mechanisms and the potential ways to combat them. Microb Pathog. 2016 Jun;95:32-42. [DOI:10.1016/j.micpath.2016.02.009] [PMID]
16. Phondani PC, Bhatt A, Elsarrag E, Horr YA. Ethnobotanical magnitude towards sustainable utilization of wild foliage in Arabian Desert. J Tradit Complement Med [Internet]. 2016;6(3):209-18. [DOI:10.1016/j.jtcme.2015.03.003] [PMID] [PMCID]
17. Qin J, Yang T, Wang H, Feng T, Liu X. Potential Predictors for Serofast State after Treatment among HIV-Negative Persons with Syphilis in China: A Systematic Review and Meta-Analysis. Iran J Public Health [Internet]. 2015 Feb [cited 2018 Sep 20];44(2):155-69. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25905049
18. Liu Y, Hardie J, Zhang X, Rotello VM. Effects of engineered nanoparticles on the innate immune system. 2018;25-32. [DOI:10.1016/j.smim.2017.09.011] [PMID] [PMCID]
19. Luo Y, Chang LW, Lin P. Metal-Based Nanoparticles and the Immune System : Activation , Inflammation , and Potential Applications. 2015;2015(Figure 1). [DOI:10.1155/2015/143720] [PMID] [PMCID]
20. Sadrieh N, Dobrovolskaia MA. Minireview: Nanoparticles and the Immune System '. 2010;151(February):458-65. [DOI:10.1210/en.2009-1082] [PMID] [PMCID]
21. Naseri N, Valizadeh H, Zakeri-Milani P. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers: Structure, Preparation and Application. Adv Pharm Bull [Internet]. 2015/09/19. 2015 Sep;5(3):305-13. [DOI:10.15171/apb.2015.043] [PMID] [PMCID]
22. Thukral DK, Dumoga S, Mishra AK. Solid lipid nanoparticles: promising therapeutic nanocarriers for drug delivery. Curr Drug Deliv. 2014;11(6):771-91. [DOI:10.2174/156720181106141202122335] [PMID]
23. Jelinkova P, Mazumdar A, Sur VP, Kociova S, Dolezelikova K, Jimenez AMJ, et al. Nanoparticle-drug conjugates treating bacterial infections. J Control Release [Internet]. 2019;307:166-85. [DOI:10.1016/j.jconrel.2019.06.013] [PMID]
24. Andrade F, Rafael D, Videira M, Ferreira D, Sosnik A, Sarmento B. Nanotechnology and pulmonary delivery to overcome resistance in infectious diseases. Adv Drug Deliv Rev. 2013 Nov;65(13-14):1816-27. [DOI:10.1016/j.addr.2013.07.020] [PMID] [PMCID]
25. Qi G, Li L, Yu F, Wang H. Vancomycin-modified mesoporous silica nanoparticles for selective recognition and killing of pathogenic gram-positive bacteria over macrophage-like cells. ACS Appl Mater Interfaces. 2013 Nov;5(21):10874-81. [DOI:10.1021/am403940d] [PMID]
26. Liu Y, Tee JK, Chiu GNC. Dendrimers in oral drug delivery application: current explorations, toxicity issues and strategies for improvement. Curr Pharm Des. 2015;21(19):2629-42. [DOI:10.2174/1381612821666150416102058] [PMID]
27. Xiong M-H, Li Y-J, Bao Y, Yang X-Z, Hu B, Wang J. Bacteria-responsive multifunctional nanogel for targeted antibiotic delivery. Adv Mater. 2012 Dec;24(46):6175-80. [DOI:10.1002/adma.201202847] [PMID]
28. Baig MS, Ahad A, Aslam M, Imam SS, Aqil M, Ali A. Application of Box-Behnken design for preparation of levofloxacin-loaded stearic acid solid lipid nanoparticles for ocular delivery: Optimization, in vitro release, ocular tolerance, and antibacterial activity. Int J Biol Macromol. 2016 Apr;85:258-70. [DOI:10.1016/j.ijbiomac.2015.12.077] [PMID]
29. Wu XT, Hong PW, Suolang DJ, Zhou D, Stefan H. Drug-induced hypersensitivity syndrome caused by valproic acid as a monotherapy for epilepsy: First case report in Asian population. Epilepsy Behav Case Reports [Internet]. 2017;8:108-10. [DOI:10.1016/j.ebcr.2017.06.003] [PMID] [PMCID]
30. Qiu Z, Yu Y, Chen Z, Jin M, Yang D, Zhao Z, et al. Nanoalumina promotes the horizontal transfer of multiresistance genes mediated by plasmids across genera. Proc Natl Acad Sci U S A. 2012 Mar;109(13):4944-9. [DOI:10.1073/pnas.1107254109] [PMID] [PMCID]
31. Lee N-Y, Ko W-C, Hsueh P-R. Nanoparticles in the Treatment of Infections Caused by Multidrug-Resistant Organisms. Front Pharmacol [Internet]. 2019;10:1153. Available from: https://www.frontiersin.org/article/10.3389/fphar.2019.01153 [DOI:10.3389/fphar.2019.01153] [PMID] [PMCID]
32. Baptista P V, Mccusker MP, Carvalho A, Ferreira DA. Nano-Strategies to Fight Multidrug Resistant Bacteria -" A Battle of the Titans ." 2018;9(July):1-26. [DOI:10.3389/fmicb.2018.01441] [PMID] [PMCID]
33. Ji Z-H, Li C-Y, Lv Y-G, Cao W, Chen Y-Z, Chen X-P, et al. The prevalence and trends of transfusion-transmissible infectious pathogens among first-time, voluntary blood donors in Xi'an, China between 1999 and 2009. Int J Infect Dis [Internet]. 2013;17(4):e259-62. [DOI:10.1016/j.ijid.2012.10.006] [PMID]
34. Li H, Chen Q, Zhao J, Urmila K. Enhancing the antimicrobial activity of natural extraction using the synthetic ultrasmall metal nanoparticles. Sci Rep. 2015 Jun;5:11033. [DOI:10.1038/srep11033] [PMID] [PMCID]
35. Lellouche J, Friedman A, Lahmi R, Gedanken A, Banin E. Antibiofilm surface functionalization of catheters by magnesium fluoride nanoparticles. Int J Nanomedicine. 2012;7:1175-88. [DOI:10.2147/IJN.S26770] [PMID] [PMCID]
36. Press D. Dual effects and mechanism of TiO 2 nanotube arrays in reducing bacterial colonization and enhancing C3H10T1 / 2 cell adhesion. 2015;(August 2013).
37. Nash KM, Ahmed S. Nanomedicine in the ROS-mediated pathophysiology: Applications and clinical advances. Nanomedicine Nanotechnology, Biol Med [Internet]. 2015;11(8):2033-40. [DOI:10.1016/j.nano.2015.07.003] [PMID] [PMCID]
38. Hu G, Guo M, Xu J, Wu F, Fan J, Huang Q, et al. Nanoparticles Targeting Macrophages as Potential Clinical Therapeutic Agents Against Cancer and Inflammation. Front Immunol [Internet]. 2019 Aug 21;10:1998. [DOI:10.3389/fimmu.2019.01998] [PMID] [PMCID]
39. Yu J, Zhang W, Li Y, Wang G, Yang L, Jin J, et al. Synthesis, characterization, antimicrobial activity and mechanism of a novel hydroxyapatite whisker/nano zinc oxide biomaterial. Biomed Mater. 2014 Dec;10(1):15001. [DOI:10.1088/1748-6041/10/1/015001] [PMID]
40. Wu B, Zhuang W-Q, Sahu M, Biswas P, Tang YJ. Cu-doped TiO(2) nanoparticles enhance survival of Shewanella oneidensis MR-1 under ultraviolet light (UV) exposure. Sci Total Environ. 2011 Oct;409(21):4635-9. [DOI:10.1016/j.scitotenv.2011.07.037] [PMID]
41. Qing Y, Cheng L, Li R, Liu G, Zhang Y, Tang X, et al. Potential antibacterial mechanism of silver nanoparticles and the optimization of orthopedic implants by advanced modification technologies. Int J Nanomedicine [Internet]. 2018 Jun 5;13:3311-27. [DOI:10.2147/IJN.S165125] [PMID] [PMCID]
42. Jiang W, Mashayekhi H, Xing B. Bacterial toxicity comparison between nano- and micro-scaled oxide particles. Environ Pollut [Internet]. 2009;157(5):1619-25. [DOI:10.1016/j.envpol.2008.12.025] [PMID]
43. Pan F, Xu A, Xia D, Yu Y, Chen G, Meyer M. Effects of octahedral molecular sieve on treatment performance , microbial metabolism , and microbial community in expanded granular sludge bed reactor Effects of octahedral molecular sieve on treatment performance , microbial metabolism , and microbial community in expanded granular sludge bed reactor. Water Res [Internet]. 2015;87(December):127-36. [DOI:10.1016/j.watres.2015.09.022] [PMID]
44. Zhang W, Li Y, Niu J, Chen Y. Photogeneration of reactive oxygen species on uncoated silver, gold, nickel, and silicon nanoparticles and their antibacterial effects. Langmuir. 2013 Apr;29(15):4647-51. [DOI:10.1021/la400500t] [PMID]
45. Mukha IP, Eremenko AM, Smirnova NP, Mikhienkova AI, Korchak GI, Gorchev VF, et al. [Antimicrobial activity of stable silver nanoparticles of a certain size]. Prikl Biokhim Mikrobiol. 2013;49(2):215-23. [DOI:10.1134/S0003683813020117] [PMID]
46. Lin J, Zhang H, Chen Z, Zheng Y. Penetration of lipid membranes by gold nanoparticles: insights into cellular uptake, cytotoxicity, and their relationship. ACS Nano. 2010 Sep;4(9):5421-9. [DOI:10.1021/nn1010792] [PMID]
47. Sarwar A, Katas H, Samsudin SN, Zin NM. Regioselective Sequential Modification of Chitosan via Azide-Alkyne Click Reaction: Synthesis, Characterization, and Antimicrobial Activity of Chitosan Derivatives and Nanoparticles. PLoS One. 2015;10(4):e0123084. [DOI:10.1371/journal.pone.0123084] [PMID] [PMCID]
48. Hyldgaard M, Mygind T, Vad BS, Stenvang M, Otzen DE, Meyer RL. The antimicrobial mechanism of action of epsilon-poly-l-lysine. Appl Environ Microbiol. 2014 Dec;80(24):7758-70. [DOI:10.1128/AEM.02204-14] [PMID] [PMCID]
49. Wehling J, Dringen R, Zare RN, Maas M, Rezwan K. Bactericidal activity of partially oxidized nanodiamonds. ACS Nano. 2014 Jun;8(6):6475-83. [DOI:10.1021/nn502230m] [PMID]
50. Foster HA, Ditta IB, Varghese S, Steele A. Photocatalytic disinfection using titanium dioxide: spectrum and mechanism of antimicrobial activity. Appl Microbiol Biotechnol. 2011 Jun;90(6):1847-68. [DOI:10.1007/s00253-011-3213-7] [PMID] [PMCID]
51. Joost U, Juganson K, Visnapuu M, Mortimer M, Kahru A, Nommiste E, et al. Photocatalytic antibacterial activity of nano-TiO2 (anatase)-based thin films: effects on Escherichia coli cells and fatty acids. J Photochem Photobiol B. 2015 Jan;142:178-85. [DOI:10.1016/j.jphotobiol.2014.12.010] [PMID]
52. Erdem A, Metzler D, Cha DK, Huang CP. The short-term toxic effects of TiO₂nanoparticles toward bacteria through viability, cellular respiration, and lipid peroxidation. Environ Sci Pollut Res Int [Internet]. 2015;22(22):17917-24. [DOI:10.1007/s11356-015-5018-1] [PMID]
53. Nataraj N, Anjusree GS, Madhavan AA, Priyanka P, Sankar D, Nisha N, et al. Synthesis and anti-staphylococcal activity of TiO2 nanoparticles and nanowires in ex vivo porcine skin model. J Biomed Nanotechnol. 2014 May;10(5):864-70. [DOI:10.1166/jbn.2014.1756] [PMID]
54. Su Y, Zheng X, Chen Y, Li M, Liu K. Alteration of intracellular protein expressions as a key mechanism of the deterioration of bacterial denitrification caused by copper oxide nanoparticles. Sci Rep. 2015 Oct;5:15824. [DOI:10.1038/srep15824] [PMID] [PMCID]
55. Yamanaka M, Hara K, Kudo J. Bactericidal actions of a silver ion solution on Escherichia coli, studied by energy-filtering transmission electron microscopy and proteomic analysis. Appl Environ Microbiol. 2005 Nov;71(11):7589-93. [DOI:10.1128/AEM.71.11.7589-7593.2005] [PMID] [PMCID]
56. Cui Y, Zhao Y, Tian Y, Zhang W, Lu X, Jiang X. The molecular mechanism of action of bactericidal gold nanoparticles on Escherichia coli. Biomaterials. 2012 Mar;33(7):2327-33. [DOI:10.1016/j.biomaterials.2011.11.057] [PMID]
57. Durmus NG, Taylor EN, Inci F, Kummer KM, Tarquinio KM, Webster TJ. Fructose-enhanced reduction of bacterial growth on nanorough surfaces. Int J Nanomedicine. 2012;7:537-45. [DOI:10.2147/IJN.S27957] [PMID] [PMCID]
58. Wang L, Hu C. The antimicrobial activity of nanoparticles : present situation and prospects for the future. 2017;1227-49. [DOI:10.2147/IJN.S121956] [PMID] [PMCID]
59. Amini Kafi-abad S, Rezvan H, Abolghasemi H, Talebian A. Prevalence and trends of human immunodeficiency virus, hepatitis B virus, and hepatitis C virus among blood donors in Iran, 2004 through 2007. Transfusion [Internet]. 2009 Oct;49(10):2214-20. [DOI:10.1111/j.1537-2995.2009.02245.x] [PMID]
60. Mohanty S, Mishra S, Jena P, Jacob B, Sarkar B, Sonawane A. An investigation on the antibacterial, cytotoxic, and antibiofilm efficacy of starch-stabilized silver nanoparticles. Nanomedicine. 2012 Aug;8(6):916-24. [DOI:10.1016/j.nano.2011.11.007] [PMID]
61. Pan F, Aihua X, Xia D, Yu Y, Chen G, Meyer M, et al. Effects of octahedral molecular sieve on treatment performance, microbial metabolism, and microbial community in expanded granular sludge bed reactor. Water Res. 2015 Dec 15;87:127-36. [DOI:10.1016/j.watres.2015.09.022] [PMID]
62. Lundberg ME, Becker EC, Choe S. MstX and a putative potassium channel facilitate biofilm formation in Bacillus subtilis. PLoS One. 2013;8(5):e60993. [DOI:10.1371/journal.pone.0060993] [PMID] [PMCID]
63. Salem W, Leitner DR, Zingl FG, Schratter G, Prassl R, Goessler W, et al. Antibacterial activity of silver and zinc nanoparticles against Vibrio cholerae and enterotoxic Escherichia coli. Int J Med Microbiol. 2015 Jan;305(1):85-95. [DOI:10.1016/j.ijmm.2014.11.005] [PMID] [PMCID]
64. Slomberg DL, Lu Y, Broadnax AD, Hunter RA, Carpenter AW, Schoenfisch MH. Role of size and shape on biofilm eradication for nitric oxide-releasing silica nanoparticles. ACS Appl Mater Interfaces. 2013 Oct;5(19):9322-9. [DOI:10.1021/am402618w] [PMID]
65. Esfandiari N, Simchi A, Bagheri R. Size tuning of Ag-decorated TiO(2) nanotube arrays for improved bactericidal capacity of orthopedic implants. J Biomed Mater Res A. 2014 Aug;102(8):2625-35. [DOI:10.1002/jbm.a.34934] [PMID]
66. Deplanche K, Caldelari I, Mikheenko IP, Sargent F, Macaskie LE. Involvement of hydrogenases in the formation of highly catalytic Pd(0) nanoparticles by bioreduction of Pd(II) using Escherichia coli mutant strains. Microbiology. 2010 Sep;156(Pt 9):2630-40. [DOI:10.1099/mic.0.036681-0] [PMID]
67. Slavin YN, Asnis J, Häfeli UO, Bach H. Metal nanoparticles : understanding the mechanisms behind antibacterial activity. J Nanobiotechnology. 2017;1-20. [DOI:10.1186/s12951-017-0308-z] [PMID] [PMCID]
68. Cha S-H, Hong J, McGuffie M, Yeom B, VanEpps JS, Kotov NA. Shape-Dependent Biomimetic Inhibition of Enzyme by Nanoparticles and Their Antibacterial Activity. ACS Nano. 2015 Sep;9(9):9097-105. [DOI:10.1021/acsnano.5b03247] [PMID]
69. Ben-Sasson M, Zodrow KR, Genggeng Q, Kang Y, Giannelis EP, Elimelech M. Surface functionalization of thin-film composite membranes with copper nanoparticles for antimicrobial surface properties. Environ Sci Technol. 2014;48(1):384-93. [DOI:10.1021/es404232s] [PMID]
70. Arakha M, Pal S, Samantarrai D, Panigrahi TK. Antimicrobial activity of iron oxide nanoparticle upon modulation of nanoparticle-bacteria interface. Nat Publ Gr [Internet]. :1-12.
71. Mehmood S, Rehman MA, Ismail H, Mirza B, Bhatti AS. Significance of postgrowth processing of ZnO nanostructures on antibacterial activity against gram-positive and gram-negative bacteria. Int J Nanomedicine. 2015;10:4521-33. [DOI:10.2147/IJN.S83356] [PMID] [PMCID]
72. Peng Y, Lo S, Ou H, Lai S. Microwave-assisted hydrothermal synthesis of N-doped titanate nanotubes for visible-light-responsive photocatalysis. J Hazard Mater [Internet]. 2010;183(1-3):754-8. [DOI:10.1016/j.jhazmat.2010.07.090] [PMID]
73. Saliani M, Jalal R. Effects of pH and Temperature on Antibacterial Activity of Zinc Oxide Nanofluid Against Escherichia coli O157 : H7 and Staphylococcus aureus. 2015;8(2). [DOI:10.5812/jjm.17115] [PMID] [PMCID]
74. Khan MF, Ansari AH, Hameedullah M, Ahmad E, Alam MM, Khan AM, et al. Sol-gel synthesis of thorn-like ZnO nanoparticles endorsing mechanical stirring effect and their antimicrobial activities : Potential role as nano-antibiotics. Nat Publ Gr [Internet]. 2016;(June):1-12. [DOI:10.1038/srep27689] [PMID] [PMCID]
75. Kazemi M, Akbari A, Feizi N. The Role of Green Reducing Agents in Gelatin-Based Synthesis of Colloidal Selenium Nanoparticles and Investigation of Their Antimycobacterial and Photocatalytic Properties.
76. Rudramurthy GR. Nanoparticles : Alternatives Against Drug-Resistant. 2016;1-30. [DOI:10.3390/molecules21070836] [PMID] [PMCID]
77. Zonaro E, Lampis S, Turner RJ, Qazi SJS, Vallini G. Biogenic selenium and tellurium nanoparticles synthesized by environmental microbial isolates efficaciously inhibit bacterial planktonic cultures and biofilms. Front Microbiol [Internet]. 2015;6:584. [DOI:10.3389/fmicb.2015.00584] [PMID] [PMCID]
78. Gordon O, Slenters TV, Brunetto PS, Villaruz AE, Sturdevant DE, Otto M, et al. Silver coordination polymers for prevention of implant infection: thiol interaction, impact on respiratory chain enzymes, and hydroxyl radical induction. Antimicrob Agents Chemother. 2010;54(10):4208-18. [DOI:10.1128/AAC.01830-09] [PMID] [PMCID]
79. Singh R, Nawale L, Arkile M, Wadhwani S, Shedbalkar U, Chopade S, et al. Phytogenic silver, gold, and bimetallic nanoparticles as novel antitubercular agents. Int J Nanomedicine. 2016;11:1889. [DOI:10.2147/IJN.S102488] [PMID] [PMCID]
80. Esmaeillou M, Zarrini G, Rezaee MA. Vancomycin capped with silver nanoparticles as an antibacterial agent against multi-drug resistance bacteria. Adv Pharm Bull. 2017;7(3):479. [DOI:10.15171/apb.2017.058] [PMID] [PMCID]
81. Lai H-Z, Chen W-Y, Wu C-Y, Chen Y-C. Potent antibacterial nanoparticles for pathogenic bacteria. ACS Appl Mater Interfaces. 2015;7(3):2046-54. [DOI:10.1021/am507919m] [PMID]
82. Saeb A, Alshammari AS, Al-Brahim H, Al-Rubeaan KA. Production of silver nanoparticles with strong and stable antimicrobial activity against highly pathogenic and multidrug resistant bacteria. Sci World J. 2014;2014. [DOI:10.1155/2014/704708] [PMID] [PMCID]
83. Payne J, Waghwani H, Connor M, Hamilton W, Dowling S, Moolani H, et al. Novel Synthesis of Kanamycin Conjugated Gold Nanoparticles with Potent Antibacterial Activity. Front Microbiol. 2016;7. [DOI:10.3389/fmicb.2016.00607] [PMID] [PMCID]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 4.0 | Iranian Journal of Medical Microbiology

Designed & Developed by : Yektaweb | Publisher: Farname Inc