Next Article in Journal
MnHR4 Functions during Molting of Macrobrachium nipponense by Regulating 20E Synthesis and Mediating 20E Signaling
Previous Article in Journal
The Deubiquitinating Enzyme USP48 Interacts with the Retinal Degeneration-Associated Proteins UNC119a and ARL3
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antifungal Effect of Nanoparticles against COVID-19 Linked Black Fungus: A Perspective on Biomedical Applications

1
Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
2
CHA Advanced Research Institute, CHA Medical Center, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12526; https://doi.org/10.3390/ijms232012526
Submission received: 23 September 2022 / Revised: 16 October 2022 / Accepted: 18 October 2022 / Published: 19 October 2022
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a highly transmissible and pathogenic coronavirus that has caused a ‘coronavirus disease 2019’ (COVID-19) pandemic in multiple waves, which threatens human health and public safety. During this pandemic, some patients with COVID-19 acquired secondary infections, such as mucormycosis, also known as black fungus disease. Mucormycosis is a serious, acute, and deadly fungal infection caused by Mucorales-related fungal species, and it spreads rapidly. Hence, prompt diagnosis and treatment are necessary to avoid high mortality and morbidity rates. Major risk factors for this disease include uncontrolled diabetes mellitus and immunosuppression that can also facilitate increases in mucormycosis infections. The extensive use of steroids to prevent the worsening of COVID-19 can lead to black fungus infection. Generally, antifungal agents dedicated to medical applications must be biocompatible, non-toxic, easily soluble, efficient, and hypoallergenic. They should also provide long-term protection against fungal growth. COVID-19-related black fungus infection causes a severe increase in fatalities. Therefore, there is a strong need for the development of novel and efficient antimicrobial agents. Recently, nanoparticle-containing products available in the market have been used as antimicrobial agents to prevent bacterial growth, but little is known about their efficacy with respect to preventing fungal growth, especially black fungus. The present review focuses on the effect of various types of metal nanoparticles, specifically those containing silver, zinc oxide, gold, copper, titanium, magnetic, iron, and carbon, on the growth of various types of fungi. We particularly focused on how these nanoparticles can impact the growth of black fungus. We also discussed black fungus co-infection in the context of the global COVID-19 outbreak, and management and guidelines to help control COVID-19-associated black fungus infection. Finally, this review aimed to elucidate the relationship between COVID-19 and mucormycosis.

1. Introduction

The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus-causing coronavirus disease 2019 (COVID-19), has led to a fourth wave of infection and immense mortality. COVID-19 can have sub-acute and prolonged long-term symptoms that can eventually damage multiple organ systems [1]. According to the World Health Organization (WHO), as of 15 October 2022, there have been 620,878,405 confirmed cases of COVID-19, including 6,543,138 deaths. As of 15 October, a total of 12,782,955,639 vaccine doses have been administered. Mucormycosis is an aggressive, severe, and rare fungal infection that can affect the patients’ pre- and post-COVID-19. Recently, post-COVID-19 infections caused by members of the order Mucorales have gained importance due to their higher incidence in immunocompromised patients with mucormycosis. They include some of the most common molds causing infections and ranked third for causing invasive infections in organ transplant and malignant tumor cases [2]. Based on the infection site, mucormycosis is clinically classified as rhino-orbital, cutaneous, pulmonary, disseminated, or gastrointestinal. Other forms can cause renal infections, osteomyelitis, endocarditis, and peritonitis and are invasive and aggressive. They can grow within blood vessels and cause thrombosis and tissue necrosis. This ability also enables hematogenous fungal dissemination [2,3].
Patients with COVID-19 are extremely prone to fungal and bacterial infections. Aspergillosis, invasive candidiasis, and mucormycosis are the most common fungal infections [4,5,6,7,8,9]. The most significant risk factors for the development of mucormycosis are diabetes mellitus, prolonged steroid use, hematological malignancy, post-stem cell transplant status, prolonged neutropenia, and impaired cell-mediated immunity [10,11]. Mucormycosis frequently affects the sinus, brain, and respiratory systems. A study on fungal cultures from 99 patients with COVID-19 found five (5%, 5/99) cases of fungal infections. These included one case of infection with Aspergillus flavus, one case with Candida glabrata, and three cases with Candida albicans [12]. Another study reported that patients with COVID-19 showed a higher percentage (8–15%) of secondary infections [13,14]. Koehler et al. [7]. reported that COVID-19-associated invasive pulmonary aspergillosis was found in 5 (26.3%) out of 19 consecutive critically ill patients with moderate-to-severe acute respiratory distress syndrome. A study from the Netherlands reported that among patients with COVID-19 who stayed in the ICU, 5 out of 31 patients were infected with Aspergillus fumigatus, and older severely ill patients were co-infected with A. flavus, as determined by tracheal aspirate culture [15,16]. Patients with COVID-19 are more susceptible to mucormycosis or other fungal co-infections due to the overexpression of inflammatory cytokines and an impaired cell-mediated immune response, with decreased CD4+ and CD8+ T cell counts. Soliman et al. [17]. reported that the toxin produced by Mucorales plays a significant role in virulence. As a result produced polyclonal antibodies against this toxin lower its ability to damage human cells in vitro, and can prevent hypovolemic shock, organ necrosis, and death in mice with mucormycosis.
The most important factors for effective antifungal therapy are early diagnosis and treatment. Moreover, surgical removal of all infected tissues and adjunctive therapies are the most effective ways to eradicate mucormycosis. For example, amphotericin B is the preferred antifungal drug for the treatment of mucormycosis. Further treatment may include triazoles, such as posaconazole, isavuconazole, and voriconazole. Recently, nanotechnology-driven innovations have provided a solution to overcome the problem of drug resistance and hence can have a profound influence on the improvement of human health. Nanotechnology-based antimicrobial agents have shown enhanced durability, performance, strength, flexibility, and unique physicochemical properties. Different types of nanoparticles have shown excellent antifungal activities and are considered a good alternative for controlling various types of fungi [18]. Specifically, metal nanoparticles can play a significant role due to their strong antifungal properties [19].
Over the past several years, the number of fungal infections has continuously increased. Over 70% of reported cases are caused by Candida spp., especially in patients with immune disorders and other serious diseases, as well as those hospitalized with serious underlying diseases [20]. Members of Candida are the most common fungal pathogens, causing diseases ranging from superficial (oral and vaginal) to systemic (such as peritonitis and meningitis candidiasis) [21]. Although a large number and diverse group of antibacterial drugs are currently available, the number of active substances for the treatment of pathogenic fungi is limited. Recent studies suggest that an increasing number of Candida infections are resistant to currently available classes of antifungal agents [22]. Hence, studying new therapeutic approaches is required for the development of novel antifungal strategies. The increased resistance of fungal pathogens to current treatments and the lack of alternative and effective treatment options are some of the greatest challenges in current medicine. Various types of therapeutic modalities, including combined drug therapy, chemical structure modifications, and the use of drug transporters, have been employed to overcome pathogenic strain resistance [23,24]. For example, several studies have found that nanoparticles can inhibit the function of efflux drug transporters, promote apoptosis of fungal cells, and reduce the formation of biofilm [25]. One study suggested that iron oxide-based nanosystems could inactivate catalase Cat-1 and thus provide a new strategy for overcoming fungal resistance. Recently, the development of nanotechnology in various applications has been rapid and immense. Nanotechnology is a new arena that focuses on particles with sizes ranging from approximately 1 to 100 nm. Nanoparticles (NPs) are widely used in nanomedicine against bacterial and fungal infections, and there is a low probability that pathogens can develop resistance to them. NPs are considered suitable candidates for transporting drugs to their targets [26,27,28]. Several types of metal and metal oxide NPs, such as silver (Ag), silver oxide (Ag2O), titanium dioxide (TiO2), zinc oxide (ZnO), gold (Au), calcium oxide (CaO), silica (Si), copper oxide (CuO), and magnesium oxide (MgO), are known to show antimicrobial activity [29]. The antimicrobial properties of nanoparticles depend on their composition, surface modification, intrinsic properties, and the type of microorganism [30]. Recently, Mal’tseva et al., reported that selenium-containing compounds such as sodium selenite, methylseleninic acid, selenomethionine, and methylselenocysteine, as well as selenoproteins and selenium nanoparticles, are involved in the regulation of defense mechanisms against various viral infections, including coronavirus infection (COVID-19) [31].
Previous studies have demonstrated that when NPs were combined with either antimicrobial peptides or antibiotics, they exhibited potential antimicrobial activity at low concentrations [32,33]. The interaction between the fungal cells and the metal is necessary for fungal resistance and homeostasis [34,35,36]. Fungi are exploited for the synthesis of nanoparticles with antimicrobial properties because they have high levels of natural metal resistance, can be economically grown, and are capable of mass production [37,38,39]. Silver is a transition metal that has properties in common with other transition metals. Silver is considered to be an effective antifungal agent for the eradication of pathogens. Silver has a positive effect on intestinal microflora, aflatoxins, and mycotoxin absorption in farm animals, and it is used in the food industry for packaging because of its antimicrobial properties [40]. Generally, Ag+ has a high affinity for sulfhydryl (thiol) and phosphate groups in enzymes required for cell wall synthesis in bacteria and fungi. In addition, it can interfere with the electron transport chain and the generation of energy. Ag+ also inhibits DNA replication and the respiratory chain in bacteria and fungi and ultimately causes cell death by generating reactive oxygen species (ROS) [41,42,43]. Gold nanoparticles (AuNPs) are generally considered to be biocompatible, non-toxic, and safer than other inorganic nanoparticles. The redox nature of Au is beneficial in reducing the ROS levels produced during exposure to NPs [44,45]. Several studies have confirmed that AuNPs exhibit significant antimicrobial activity against various clinical strains of Candida [34,46].
Zinc oxide nanoparticles (ZnO NPs) are more active antimicrobial agents against various types of microbes than other NPs in the same group of elements. Because ZnO NPs have potential UV-absorbing properties and high catalytic efficiency, they have been widely used in cosmetic lotions and as successful UV blockers. They have also been used as semiconductors in microelectronic devices and for accelerating the degradation of water pollutants by photocatalytic activity [47]. ZnO NPs exhibited enhanced cytotoxicity by increasing the production of ROS, which leads to oxidative stress and eventual cell death [48,49]. ZnO NPs had significant inhibitory effects on the growth of the C. albicans fungus. These NPs inhibit the growth of C. albicans by directly contacting the fungal cell membrane and affecting its interaction with the cell wall which can prevent growth and lead to cell death [50]. The exposure of fungal cells to NPs reduces the effectiveness of their anti-oxidative mechanisms and oxidative enzymes, and also disrupts their osmotic balance and cellular integrity. Carbon-based nanoparticles, such as fullerenes, single-walled carbon nanotubes (SWCNTs), graphene oxide (GO), and reduced GO, also exhibit antimicrobial activity (Gurunathan et al., 2012) [51]. Carbon-based nanomaterials cause membrane damage in microorganisms due to oxidative stress. The interactions between microbial cells and carbon-based nanomaterials play a significant role in their antimicrobial activity. Carbon nanomaterials can come into direct contact with the cells, which causes cell death [52,53,54,55]. Copper is an important biocompatible element involved in various physiological processes and has been effectively used to eradicate microorganisms.
Titanium dioxide (TiO2) is an inorganic NP that is non-toxic, antibacterial, and super lipophilic. It also promotes bacterial decomposition and protects against ultraviolet (UV) rays [56,57]. Because of these factors and its broad range of functions, TiO2 is among the most widely used NPs in industry and consumer applications. There are three forms of TiO2 crystal structures: anatase, rutile, and brookite [58]. TiO2 NPs exhibit potential antimicrobial activities as observed against C. albicans in the presence or absence of sunlight. Iron oxide NPs (IONPs) are considered safe and non-toxic and are widely used because of their magnetic and biochemical properties as well as their low cost of use in various medical applications. IONPs have been shown to have broad-spectrum antiviral [59], antiparasitic [60], bactericidal [61,62], antibiofilm, antifungal [63], and wound healing properties [64]. IONPs have attracted immense interest due to their remarkable physiochemical properties and have been utilized in a broad range of applications, including biomedicine, electronics, catalysis, water treatment, and magnetic and environmental remediation [65,66].
Several studies have reported the development of severe opportunistic infectious diseases, such as pneumonia, candidiasis, and pulmonary aspergillosis, in patients with COVID-19 [67]. Mucormycosis is an acute and invasive fungal infection caused by Mucorales-related fungal species, and it is significantly more likely to occur in immunocompromised patients. Mucormycosis infections are caused by ubiquitous opportunistic fungi belonging to the order Mucorales, family Mucoraceae, and they are commonly known as the black fungus [68,69]. The novel SARS-CoV-2 causes susceptibility to two different fungal infections, COVID-19-associated pulmonary aspergillosis (CAPA) and COVID-19-associated mucormycosis (CAM) [70]. One study found that more than 47,000 cases of CAM were reported in merely three months in India [71]. Patients with diabetes are at a higher risk of infection with COVID-19-linked black fungus because of their high levels of blood sugar. Researchers believe that high blood sugar levels make the body a more conducive environment for the growth of fungi. Another risk factor is the use of corticosteroids, which are strong immunosuppressants used to treat severe cases of COVID-19 [71,72]. The mortality rate was significantly increased in COVID-19 patients with black fungus infection. Among COVID-19-infected countries, the mortality rate of black fungus-infected patients was significantly higher in India [73]. The symptoms of mucormycosis, such as reddish swollen skin over the nose and around the eyes, have been observed. Other symptoms include vision abnormalities, eye swelling, ocular pain, facial edema, and shortness of breath [74]. Rapid diagnosis of mucormycosis can be performed using fluorescent brighteners. Furthermore, fungal culturing techniques allow identification at the genus and species levels. Quantitative PCR is a very sensitive and rapid method to diagnose mucormycosis in patients who cannot undergo a biopsy or in patients with hematological malignancies due to severe thrombocytopenia [75,76].
This review presents a wide-ranging and detailed analysis of the current progress and challenges in the application of metal nanoparticles for controlling COVID-19-associated black fungus infection. Particularly, we discuss the effects of nanoparticles on fungal infections and the use of various types of metal nanoparticles as potential antifungal agents against different types of fungi. Further, we discuss the pathogenesis of mucormycosis, and the effect of nanoparticles on black fungus, and also we discuss black fungus co-infection in the context of the global COVID-19 outbreak. Then, we discuss the management and guidelines to control COVID-19-related black fungus infection. Finally, we present our conclusions and possible directions for further research.

2. Silver Nanoparticles

Engineered silver nanoparticles (AgNPs) are some of the most common nanoproducts used against microorganisms [28,77,78,79]. The toxic nature of AgNPs depends on their size, composition, surface area, charge, redox potential, and concentration [80,81]. When the fungus Arthroderma fulvum strain HT77 was used to biologically synthesize AgNPs, the resulting nanoparticles exhibited antifungal activity against a variety of fungal pathogens, including Candida spp., Aspergillus spp., and Fusarium spp. Pd@Ag nanosheets (Pd@Ag NSs) also exhibited excellent inhibitory antifungal activity against common invasive fungal pathogens, such as those in the Cryptococcus neoformans species complex. This included fluconazole-resistant isolates. The anticryptococcal activity of the Pd@Ag NSs was significantly greater than that of fluconazole and similar to that of amphotericin B (AmB). Pd@Ag NSs exhibited fungicidal activity against Cryptococcus spp. by disrupting cell integrity, intracellular protein synthesis, and energy metabolism [28]. AgNPs were found to have potent antifungal activity against Candida by generating ROS. They also affect other cellular targets, resulting in alterations in membrane fluidity, cellular morphology, and ultrastructure t as well as ergosterol content [82]. AgNPs synthesized from crude bioactive metabolites of bionanofactories isolated from Lake Mariout exhibited antibacterial, antifungal, and antibiofilm activities. Silver/chitosan nanocomposites with an average size of 10 nm have the potential to inhibit the growth of Penicillium expansum, which is the causative agent of blue mold-contaminated dairy cattle feed [83]. AgNPs synthesized from aqueous pigeon dropping (PD) extract inhibited various types of microorganisms, such as Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Bacillus spp. They also showed the highest antifungal effect against A. flavus and the lowest effect against Penicillium griseofulvum. Das et al. [84]. reported that the surface coating of biogenic AgNPs with zinc oxide increased its antimicrobial efficiency against Candida krusei. AgNPs produced by propolis extract-mediated synthesis exhibited antifungal activities against various fungi, including Candida sp., Fusarium sp., Microsporum sp., and Trichophyton sp. AgNPs also acted synergistically with fluconazole to inhibit the growth of fluconazole-resistant C. albicans by abrogating drug efflux pumps and increasing endogenous ROS production. Treatment with AgNPs downregulated ERG1, ERG11, ERG25, and CDR2, decreased membrane ergosterol levels and membrane fluidity. It also reduced the membrane content of Cdr1p and Cdr2p, and thus efflux pump activity. A combination of Ag-, Au-, metallic- and ZnO-NPs effectively controlled the growth of four mycotoxin-producing mold strains, including A. flavus and A. fumigatus [85]. Treatment with AgNPs significantly inhibited the growth of six fungal species, including C. albicans, C. glabrata, Candida parapsilosis, C. krusei, and Trichophyton mentagrophytes by altering the structure of the cell membrane and inhibiting the normal budding process [86,87,88]. Monodispersed monometallic and bimetallic nanoparticles, which are highly reactive NPs with sizes less than 5 nm, exhibited high antifungal activity against C. parapsilosis, C. krusei, C. glabrata, Candida guilliermondii, and C. albicans [89]. Immobilized AgNPs on chitosan also enhanced antifungal efficacy against Monilia albicans [90]. In addition, a nanocomposite containing pullulan and AgNPs exhibited antifungal activity against Aspergillus niger [91] Several studies demonstrated that AgNPs can act as potential antifungal agents, although this depends on their sizes, shapes, and zeta potentials. Moreover, the mechanism of inhibition of fungi varies with particle size. AgNPs were found to be effective against various fungal pathogens, including C. albicans, Candida tropicalis, Trichophyton rubrum, Penicillium brevicompactum, Cladosporium cladosporioides, A. fumigatus, Chaetomium globosum, Mortierella alpina, and Stachybotrys chartarum [19,92,93,94]. A different study was conducted to determine the efficacy of AgNPs against various fungal pathogens that cause fungal keratitis, a vision-threatening infection. AgNPs exhibited antifungal activity with minimal inhibitory concentrations of 1, 0.5, and 0.5 μg/mL for Fusarium spp., Aspergillus spp., and Alternaria alternata, respectively [95]. Increasing the concentration of Ag-embedded mesoporous silica nanoparticles (mSiO2@AgNPs) using a leaf extract of Azadirachta indica improved its potential antifungal activity against C. albicans [96]. AgNPs functionalized with phenolic compounds also showed potential antifungal effects against A. niger by interfering with spore germination and mycelial growth. Biologically synthesized ultrasmall silver nanoclusters (rsAg@NCs) using metabolites from usnioid lichens exhibited excellent antimicrobial activity against fluconazole (FCZ)-resistant C. albicans, which was significantly higher than that of chemically synthesized silver nanoparticles (AgNPs) and FCZ. The rsAg@NCs induced apoptosis via ROS generation, which leads to the loss of mitochondrial membrane potential, DNA fragmentation, chromosomal condensation, and metacaspase activation [97]. AgNPs synthesized by Mucor hiemalis showed significant antibacterial activity against six pathological bacterial strains (Klebsiella pneumoniae, Pseudomonas brassicacearum, Aeromonas hydrophila, E. coli, Bacillus cereus, and S. aureus) and three pathological fungal strains (C. albicans, Fusarium oxysporum, and A. flavus). AgNPs were found to synergistically inhibit bacterial growth when used in combination with several antibiotics (streptomycin, tetracycline, kanamycin, and rifampicin). In addition, they were found to inhibit the growth of fungi when used in conjunction with several fungicides (amphotericin B, fluconazole, and ketoconazole). AgNPs synthesized using aqueous Eucalyptus camaldulensis showed strong fungicidal activity ranging from 0.5 to 1 µg/mL affecting Candida adhesion and invasion into host cells by reduced germ tube formation and hydrolytic enzyme secretion. Furthermore, AgNPs downregulated the expression of ALS3, HWP1, ECE1, EFG1, TEC1, and ZAP1, which encode proteins required for hyphal growth and biofilm development, and PLB2, LIP9, and SAP4, which encode hydrolytic enzymes [98]. AgNPs synthesized using Cinnamomum camphora fruit extract exhibited excellent antifungal effects against F. oxysporum by inhibiting colony growth and conidia germination [13]. The metal nanoparticles (MNPs) synthesized by Fusarium solani showed also showed potential antifungal effects against multidrug-resistant (MDR) P. aeruginosa and S. aureus in addition to its mycotoxigenic activity against Aspergillus awamori, A. fumigatus, and F. oxysporum [99].

3. Zinc Oxide Nanoparticles

Fungal pathogens present a major health problem and can cause severe damage. Chitosan (CS) functionalized with linoleic acid (LiA) quantum dots inhibited the growth of fluconazole-resistant clinical strains at concentrations similar to those of fluconazole [100]. ZnO NPs inhibit the viability of C. albicans in a concentration-dependent manner. The minimal concentration of ZnO NPs (0.1 mg/mL) inhibited the growth of C. albicans by over 95%. The loss of viability was reduced in the presence of histidine, suggesting the involvement of reactive oxygen species, including hydroxyl radicals and singlet oxygen, in cell death [101]. Nano-ZnO doped with 5% nano-Pd, pure nano-ZnO, and micro-ZnO, showed antifungal activity against A. niger with minimum inhibitory concentrations (MICs) of 1.25, 2.5, and 5 mg/mL, respectively. Among the three tested compounds, nano-ZnO exhibited a significantly stronger induction of cell death in Aspergillus compared to its non-nano-counterpart [102]. ZnO QDs at concentrations higher than 3 mmol/L inhibited the growth of P. expansum [103]. ZnO NPs embedded within a mesoporous nanosilica (mSiO2) matrix (ZnO@mSiO2) inhibited the growth of four strains of fungi in a dose-dependent manner. ZnO NPs remarkably reduced germ tube formation in C. albicans by decreasing phospholipase and proteinase secretion. ZnO NPs penetrate the cell and cause extensive damage to the cell wall and cell membrane [104]. Fungal keratitis (FK) is caused by pathogenic bacterial and fungal strains that contaminate contact lenses. Zinc oxide nanoparticles functionalized with gallic acid (GA) exhibited antifungal activity against various types of fungal strains, including C. albicans, A. fumigatus, and F. solani [105]. The growth inhibition of Penicillium oxalicum, Pestalotiopsis maculans, Paraconiothyrium sp., and A. niger fungi achieved by Mg1x ZnxO QDs was higher than that obtained by either of the two pure oxide QDs [106]. ZnO QDs time- and dose-dependently inhibited radial growth against two different types of fungi, such as A. fumigatus and C. albicans [107]. Cierech et al. [108]. reported the antifungal properties of PMMA and doped ZnO NPs were mediated by the inhibition of biofilm deposition on the surface in a concentration-dependent manner. Syzygium aromaticum functionalized ZnO NPs (SaZnO NPs) reduced the growth and production of deoxynivalenol and zearalenone in Fusarium graminearum through the accumulation of ROS in a dose-dependent manner. Furthermore, SaZnO NPs treatment enhanced lipid peroxidation, depleted ergosterol content, and caused detrimental effects on fungal membrane integrity [109].

4. Gold Nanoparticles

Gold nanoparticles (AuNPs) have unique properties and are not inherently toxic to human cells, which makes them promising therapeutic agents [110,111,112]. AuNPs have been widely used in cancer treatments as a drug delivery system and in thermal therapy [113,114]. In addition, AuNPs have shown potential activity against various pathogens, including E. coli, Streptococcus mutans, and Candida species [34,115,116]. Khan et al. [117]. reported that antibiofilm assays and microscopic studies showed that gold nanoparticle-methylene blue conjugates significantly reduced biofilm formation [117]. The shape of nanoparticles plays a significant role in their antimicrobial activity. To demonstrate the impact of the shape of the silver and gold nanoparticles, nanoparticles of different sizes were selected, including nanocubes, nanospheres, and nanowires, and their effect on C. albicans, C. glabrata, and C. tropicalis was assessed. The study found that silver and gold nanocubes had higher antifungal properties against the test species than did nanospheres or nanowires [118]. AuNPs directly interacted with DNA and led to nuclear condensation and DNA fragmentation. AuNPs can cause an overload of mitochondrial Ca2+ and the collapse of mitochondrial Ca2+ homeostasis. Thus, the disruption of mitochondrial Ca2+ homeostasis could play a role in initiating AuNP-induced cell death in C. albicans [119]. AuNPs have been shown to strongly inhibit pathogenic biofilm formation and the invasion of dental pulp stem cells (DPSCs). Moreover, treatment with AuNPs led to the activation of immune response-related genes in DPSCs, which could enhance the activity of the host immune system against pathogens [120]. Antimicrobial activity was evaluated using stabilized cationic dipeptide-capped gold/silver nanohybrids, in which peptide-capped AgNPs exhibited higher antimicrobial activity than peptide-capped AuNPs, native peptides, or unconjugated metallic nanoparticles [121]. AgNPs and AuNPs were synthesized using sodium borohydride and tannic acid as reducing agents, and then both were tested for antifungal activity against C. albicans and Saccharomyces cerevisiae. Of the two reagents, AgNPs showed greater potential antifungal activity compared to that of AuNPs [122]. Quercetin-functionalized AuNPs showed significant antioxidant activity compared to quercetin alone. It also exhibited significant antifungal activity against A. fumigatus at concentrations ranging from 0.1 to 0.5 mg/mL [123]. Heparin-functionalized gold nanoparticles (AuHep-NPs) with an average size of 530 nm also showed significant antifungal activity against C. albicans, Issatchenkia orientalis (C. krusei), and C. parapsilosis [124]. AuNPs coated with the antimicrobial peptide indolicidin inhibited biofilm formation and development in clinical isolates of Candida spp. [46]. A further study suggested that indolicidin-conjugated AuNPs exhibited potential antifungal effects against fluconazole-resistant strains of C. albicans isolated from patients with infected burns [125].

5. Copper Oxide Nanoparticles

Copper has been widely used as an antimicrobial agent. Several studies have demonstrated the antimicrobial activity of copper nanoparticles (CuNPs) against multiple fungi [126,127,128]. Oussou-Azo et al. [129]. reported that all the forms of copper they studied inhibited hyphal growth in a dose-dependent manner. They also reported that CuNPs and CuO NPs showed the greatest inhibitory effects against fungal pathogens. Cu NPs are considered effective in inhibiting the growth and colony formation of Colletotrichum gloeosporioides. The toxicity of copper particles depends on a combination of several factors, such as concentration, length of exposure, humidity, and temperature. Copper induces cell death by interacting with microorganisms, and it can alter cell membrane permeabilization, membrane lipid peroxidation, protein alteration, and denaturation of nucleic acids, which ultimately leads to cell death. Surface coatings of copper are also able to provide antifungal effects [30,130,131,132]. The CuO NPs exhibited maximum antibacterial and antifungal activities against human clinical pathogens by the generation of free radicals, especially nitric oxide radicals [133]. In addition, decorating Ag NPs with binary silver/copper nanoparticles increased the antibacterial and antimycotic activity of cotton fabrics. Textile materials containing ANPs demonstrated high antibacterial activity, whereas fabrics doped with bimetallic composite Ag/Cu had pronounced antimycotic properties [134]. CuO NPs with a spherical shape and an average size between 20 and 50 nm were shown to exert antifungal activity against Fusarium sp. at a concentration of 450 ppm. After a nine-day incubation at this concentration, 93.98% of fungal growth was inhibited [135]. Green synthesis of CuNPs resulted in CuNPs that had antifungal effects against F. solani, Neofusicoccum sp., and F. oxysporum. These CuNPs acted by damaging the cell membranes of the pathogens and by inducing the intracellular generation of ROS in the mycelium [136]. CuO NPs synthesized using Cissus quadrangularis plant extract had an average size of 30 ± 2 nm. The synthesized CuO NPs exhibited dose-dependent potential activity against A. niger [137]. Copper oxide nanocomposites (CuO/C nanocomposites) with a particle size of 50 nm have also been prepared using sucrose as a capping agent. CuO/C nanocomposites exhibited potential antifungal activity against A. niger and A. flavus species [138]. CuNPs with a size range of 23–82 nm and a round to polygonal shape also exhibited potential antifungal activity against rotting plant pathogens, such as F. oxysporum and Phytophthora parasitica [139]. CuO NPs synthesized using green and black tea leaf extracts containing flavonoids and phenols that have a spherical shape and an average size between 26 and 40 nm also exhibited antibacterial and antifungal activities. Synthesizing CuO NPs using Persea americana-derived seed extracts that contained carboxylic acid alkanes resulted in CuO NPs with a spherical shape and an average size from 42 to 90 nm. These CuO NPs also showed antibacterial, antifungal, and antioxidant activities [140]. Microwave-mediated synthesis of CuO NPs resulted in CuO NPs that were spherical in shape and had an average diameter of 7–10 nm and that showed significant antifungal activity against Cladosporium herbarum [141]. Aqueous-phase mediated synthesis of nanoparticles, such as copper/copper oxides, also resulted in NPs that showed antifungal effects against F. oxysporum. Both of these nanoparticles showed a high percentage of inhibition of radial growth (IGR) [142]. CuO NPs significantly induced cell death in C. albicans by stimulating the production of ROS and by causing membrane damage. This damage eventually leads to the suppression of yeast-to-hyphae morphological switching by downregulating cph1, hst7, and ras1 and by upregulating the negative regulator tup1 [143]. Martínez et al. [144]. evaluated the in vitro activity of CuNPs and copper nanowires against C. albicans strains and yeast. Microscopic analysis revealed that treatment with CuNPs and nanowires altered cell morphology and cell wall, which ultimately leads to the complete collapse of the yeast. When C. albicans was exposed to CuNPs and nanowires, the nanoparticles and nanowires exhibited potential antifungal activity by releasing free Cu2+ ions that acted as a biocide. In addition, the sharp edges of marigold-like petal nanostructures were able to injure the cell wall and membrane and cause the death of the yeast. The starch-and sodium alginate-mediated synthesis of CuNPs resulted in nanoparticles that showed antifungal activities against C. albicans and C. krusei [145]. Silver and CuO NPs significantly reduced the mycelial growth of A. alternata and P. oryzae in a dose-dependent manner [146].

6. Titanium Dioxide Nanoparticles

Titanium dioxide nanoparticles (TiO2 NPs) have a wide range of applications in industry and medicine, including in drug delivery systems and water purification. TiO2 NPs have unique optical, electronic, photocatalytic, and antimicrobial properties [147]. Candida species are known to cause serious cutaneous, mucosal, and systemic fungal infections. TiO2 NPs in their anatase and rutile forms displayed significant antifungal activity against C. albicans. The loss of cell viability was dose- and time-dependent. TiO2 NPs also alter cell morphology [148]. Due to their high photocatalytic activity, photocatalytic paints formulated with the anatase form of TiO2 exhibited antifungal effects against A. niger. Several studies have reported the antifungal activity of photocatalytic paints using A. niger as an environmental model, along with non-pathogenic molds [149,150]. Moradpoor et al. [151]. reported that TiO2 NPs exhibited significant antifungal activities against oral C. albicans pathogens. N and F co-doped TiO2 nanoparticles with sizes of 200–300 nm were used as antifungal agents under visible light. The colloidal form, with its appropriate surface chemistry, allows the efficient interaction of the nanoparticles with the fungal cell wall, and visible light-induced ROS generation by the nanoparticles results in fungal disinfection [152].
A study was conducted to compare the antifungal activities of TiO2 NPs and ZnO NPs with those of amphotericin B (AmB) against different species of Candida. The results showed that TiO2 and ZnO NPs exhibited antifungal activity against pathogenic Candida species. The results revealed that TiO2 and ZnO NPs were significantly less active than AmB [153]. In the same study, antifungal activity was evaluated using various types of nanoparticles, such as ZnO, TiO2, and ZnO–TiO2, by the sol-gel method. All synthesized nanoparticles (50 μg/mL) showed fungal growth inhibition, and ZnO–TiO2 showed higher antifungal activity against A. flavus than pure TiO2 or ZnO. In combination, ZnO and TiO2 showed potential antifungal activity at a low concentration (150 μg/mL) of ZnO–TiO2 and exhibited higher ROS production and induction of oxidative stress [154]. UV-irradiated TiO2 NPs attack microbial phospholipids, thereby causing cell damage [155]. TiO2 released ROS upon direct contact with the cell membranes of bacterial cells. TiO2 NPs have a positive charge on their surface, and microbial cell membranes have a negative charge, which enhances their attachment [156]. TiO2 NPs show a high affinity for membrane proteins and can alter their structures. Photocatalysis also influences the ability of TiO2 NPs to bind with phosphoproteins and phospholipids [157,158]. Under UV light, the electron holes in nano-TiO2 interact with water and oxygen to generate active oxygen species, especially hydroxyl radicals, thereby achieving antimicrobial effects [159]. TiO2-coated film significantly inhibited the development of Penicillium rot in apples through a photocatalytic reaction and reduced the risk of microbial growth on fresh-cut produce [160,161]. Chitosan nanoparticles detrimentally affect the function of the cell membrane and exhibit antibacterial properties. Increasing the concentration of chitosan leads to the degradation of chitin, thus destroying the cell wall of the pathogen and achieving an antifungal effect [162,163]. Chitosan and TiO2 composites can adsorb onto the cell surface well and easily penetrate the cell wall, thereby causing the leakage of intracellular material and impaired nuclear function. this results in the inhibition of cell growth or death. Similarly, the oxygen free radicals produced can attack the outer membrane, DNA, RNA, and lipids. This oxidation can damage bacteria, leading to the inhibition of cell growth or death [164,165,166]. Chitosan can alter the permeability of a fungal cell membrane, resulting in extensive electrolyte leakage and, consequently, cell death [167]. Nano-TiO2 treatment produces strong oxides, such as hydroxyl radicals, which can destroy the unsaturated bonds in organic molecules, damage cell components, break the cell wall membrane, and destroy the permeable barrier, all of which ultimately result in cell death [168,169].

7. Magnetic and Iron Oxide Nanoparticles

Magnetic nanoparticles of iron oxides (Fe3O4 and/or γ-Fe2O3) are currently used as contrast agents in magnetic resonance imaging (MRI) studies and cancer treatments [170]. Nehra et al. [171]. investigated the antibacterial and antifungal activities of bare and chitosan-coated Fe3O4 NPs against five organisms. The results showed that chitosan-coated Fe3O4 NPs could be potentially effective antimicrobial agents against various microorganisms, such as E. coli, Bacillus subtilis, C. albicans, A. niger, and F. solani. Biologically synthesized Fe2O3-CsNPs significantly inhibit the Verticillium wilt pathogen Verticillium dahlia by altering the morphology of its hyphae [172]. A study was performed to evaluate the antifungal activity of IONPs against different species of Candida. compared with fluconazole (FLC). The results suggested that IONPs can potentially inhibit the growth of C. tropicalis, C. albicans, and C. glabrata [173]. One study reported that incubation of fungal cells with MNPs inactivates the catalase Cta1 and disturbs the oxidation-reduction balance [174]. Furthermore, studies suggest that MNPs coated with LL-37 or CSA-13 were able to increase the generation of ROS, which is probably associated with the mechanism of action of CAPs, which results in pore formation within cell membranes. The formation of these pores, contributes to nanoparticle transport into Candida cells. Excessive ROS production can destroy the integrity of the fungal cell wall, which enhances MNP internalization and oxidative damage [175]. Unmodified nanoparticles strongly inhibit C. albicans biofilm formation due to cell wall disruption, resulting in the loss of fungal biofilm structure [176]. Magnetite nanoparticle-decorated reduced graphene oxide showed antifungal activities against T. mentagrophytes and C. albicans. Hence, rGO/Fe3O4 NCs are used as excellent adsorbents to remove organic dye pollutants and kill pathogens.
IONPs are of immense interest due to their superparamagnetic properties and their potential biomedical applications, which arise from their biocompatibility and non-toxicity [177]. IONPs have various applications, including drug and gene carriers for target-specific drug delivery and gene therapy, respectively. The surface coating of IONPs not only decreased their cytotoxicity, but also increased the stability and efficiency of the antimicrobial potential of IONPs [178]. IONPs synthesized using P. orientalis showed significant antifungal activities against Mucor piriformis as compared with A. niger [179]. Salari et al., (2018) evaluated the biofilm formation ability of different Candida strains and the antibiofilm effects of Fe3O4-NPs compared with fluconazole (FLC). After exposure to various concentrations of Fe3O4-NPs, the ability of the nanoparticles to reduce biofilm formation in C. albicans and C. parapsilosis was greater than that of FLC. The iron oxide nanoparticles (Fe2O3-NPs) in the study had sizes of 10–30 nm and were prepared using a green approach that used tannic acid as a reducing and capping agent. The effect of the prepared nanoparticles was evaluated using Trichothecium roseum, C. herbarum, Penicillium chrysogenum, A. alternata, and A. niger. Among the tested fungi, Fe2O3-NPs were found to be the most effective against P. chrysogenum and A. niger. Fluconazole (FLZ) on chitosan (CS)-coated IONPs potentially inhibits Candida planktonic cells [180].

8. Carbon Nanoparticles

Graphene oxide (GO) is a new nanomaterial with different potential biomedical applications due to its excellent physicochemical properties and ease of surface functionalization. Carbon nanoscrolls (CNSs) filled with AgNPs exhibited ideal lengthened activities against C. albicans and C. tropicalis when compared with the GO-AgNPs nanocomposites [181]. Cationic fullerene derivatives bearing a substituted-quinazolin-4(3H)-one moiety exhibited significant activity against various fungal strains, including C. albicans, A. clavatus, and A. niger [182]. Polyethelene glycol functionalized GO potentially inhibits cell viability and can control fungal infections by limiting the growth of the opportunistic fungal pathogen C. albicans and activating immune effector cells. Thus, it can prevent invasive candidiasis [183]. Size-controllable red-emissive carbon dots (CDs) with positively charged guanidine groups (Gu+) and conjugated with the antifungal polyene AmB exhibited potent antifungal effects in both the planktonic and biofilm forms [184]. The antibacterial and antifungal activity of CDs derived from tamarind arises from its interaction with DNA via intercalation [185]. Their antifungal activity against C. albicans was evaluated using a novel hydrophilic nanoconjugate gold@carbon dot (Au@CD), among others. The nanoconjugates exhibited a profound effect on the susceptibility and size of C. albicans cells [186]. Reduced graphene oxide (rGO) inhibited the mycelial growth of several fungal pathogens, including A. niger, Aspergillus oryzae, and F. oxysporum [187]. Single-walled carbon nanotubes (SWCNTs), multi-walled carbon nanotubes (MWCNTs), fullerene (C60), and rGO all show significant antifungal activity against F. graminearum and Fusarium poae [188]. Graphene sheets also showed potential antifungal effects by disturbing fungal structures and damaging the cell wall with their sharp edges (Xie et al., 2016) [189], producing oxidative stress [190]. Nanoparticles inhibit fungal growth by entering the cells through endocytosis and diffusion processes, and their entry will eventually lead to apoptosis. The mechanism of antifungal activity of various types of NPs includes five different steps: (1) internalization of NPs through endocytosis and diffusion process, (2) disassembly of the membrane, (3) release of nanoparticles into the cytoplasm, (4) inducing oxidative stress, and (5) ROS induced cell death by the activation of pro-apoptotic proteins and ROS production, which leads to mitochondrial dysfunction and the endoplasmic reticulum stress-mediated apoptosis pathway (Figure 1). Miragoli et al., (2013) reported that positively charged, amine-modified polystyrene latex nanoparticles induced large-scale damage to cardiomyocyte membranes leading to calcium alternans and cell death. Conversely, negatively charged, carboxyl-modified polystyrene latex nanoparticles (NegNPs) triggered the formation of 50–250-nm nanopores in the membrane. In vitro study demonstrated that when ventricular myocytes were exposed to TiO2 they had significantly reduced action potential duration, impairment of sarcomere shortening, and decreased stability of resting membrane potential [191]. Furthermore, in vivo studies depicted that TiO2 nanoparticles increased cardiac conduction velocity and tissue excitability, resulting in an enhanced propensity for inducible arrhythmias [192]. An antifungal effect of representative nanoparticles is summarized in Table 1 [136,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214].
Mucormycosis is caused by various saprophytic fungi in the order of Mucorales, including members of the Rhizopus, Lichtheimia, Mucor, and Rhizomucor genera [215]. These fungi affect various organs, including the lungs, skin, gut, rhinocerebral areas, and central nervous system [216]. Rhizopus arrhizus is the most common causative agent globally, and Apophysomyces is the second most common species to cause mucormycosis [217]. Generally, these fungi are relatively larger than other species and thus are easily retained in the paranasal sinuses. However, certain smaller species may also reach the lungs [218]. Usually, the spores of these fungi disperse through the air and enter humans via inhalation, inoculation in skin lesions and other similar areas, and orally, which allows them to enter the gastrointestinal tract [219]. Fungal propagation involves spore inoculation, escaping phagocytosis by macrophages and neutrophils, hyphae germination, growth by taking advantage of the condition of the host (such as an iron overload or ketoacidosis), attaching to the endothelium using receptors, and damaging the endothelium. This process results in tissue necrosis, thrombus formation, or hemorrhage (Figure 2). All of these conditions contribute to multiorgan dysfunction. The spores enter into the human body by inhalation, ingestion, or direct inoculation through wounds, and then germinate angioinvasive hyphae [220]. Fungi enter the human body through adherence to the extracellular matrix proteins present in the basement membrane, laminin, and collagen VI by specific binding and subsequently secrete lipolytic/glycosidic enzymes and proteases that degrade the underlying stroma, ultimately facilitating fungal invasion into the host tissues [221,222].
The pathogenesis of mucormycosis includes the process of thrombosis, in which the fungi adhere to the endothelial cells and disrupt their integrity so as to gain access to the bloodstream [223]. Fungal spores bind to coat homolog proteins on their surface which enables the spores to effectively bind by means of adhesins to 78-kDa glucose regulator protein 78 (GRP78) receptors present in the endothelial cells [222]. Upregulation of GRP78, a heat shock protein due to increased glucose and iron content caused by diabetic ketoacidosis (DKA) or induced by the use of dexamethasone, also facilitates fungal entry [224]. This is especially true for patients with COVID-19. After binding to the blood vessels, secondary metabolites released during the phagocytosis of R. oryzae by the endothelial cells are the cause of the disruption to the endothelial lining, and not viable fungi [225]. Hyperglycemia is a significant risk factor for mucormycosis. COVID-19 infection may directly induce hyperglycemia by damaging pancreatic beta cells or by the use of corticosteroid therapy, which may contribute to CAM pathogenesis. GRP78 receptors not only play a role in the pathogenesis of COVID-19 but are also involved in mucormycosis pathogenesis. Increased expression of the GRP78 receptor may also enhance the binding of Mucorales spores, resulting in enhanced endothelial invasion and damage in patients with COVID-19 [226,227].
Mucormycosis significantly affects patients with COVID-19 in three ways. COVID-19 leads to a reduction in the availability of T cells, an increase in pro-inflammatory markers in patients with severe disease, and pronounced damage to the pulmonary tissues. All of these changes aid in fungi invasion [228,229]. The cytokine storm caused by COVID-19 increases the level of IL-6, which can lead to high levels of iron and macrophage activation. This can result in the increased availability of free iron within cells. These free iron species support the growth of fungi. This, in turn, causes endothelial destruction and inflammation, which is called endothelitis [224]. Thus, COVID-19–induced immunosuppression increases the risk of opportunistic infection, damages the endothelium and alveolae (which makes fungal invasion easier), and increases the glucose level due to the acute diabetes-like state caused by pancreatic damage. The increased ferritin level also supports fungal growth, and an increase in body temperature is optimal for thermotolerant fungi. In patients with uncontrolled diabetes and poor glycemic control, ketone concentrations in the blood increase, which leads to acidosis [227,230].
Iron plays a significant role in mucormycosis infection in patients with COVID-19. Iron-binding proteins, such as transferrin, ferritin, and lactoferrin, limit the release of free iron in the blood and the maintenance of iron homeostasis [231]. Patients with diabetes mellitus show increased serum ferritin levels, which leads to increased iron stores [232]. In patients with COVID-19, SARS-CoV-2 interacts with the hemoglobin molecule and causes the dissociation of iron from heme molecules, which leads to hyperferritinemia in patients with COVID-19 [233]. Increased levels of serum ferritin, IL-6, and D-dimers have been associated with high mortality in patients with COVID-19 [234]. Hyperferritinemia in patients with COVID-19 leads to increased lung inflammation and lung fibrosis, leading to severe disease [235]. Free iron in the blood facilitates the growth and invasion of blood vessels, causing vessel thrombosis and tissue necrosis [219].

9. Impact of Nanoparticles on Black Fungus

Coronaviruses are a diverse group of viruses that infect many different animals and can cause mild-to-severe respiratory infections in humans. Among the various types of coronaviruses, SARS-CoV-2 and Middle East respiratory syndrome coronavirus (MERS-CoV) caused pathogenic diseases in 2002 and 2012, respectively [236]. COVID-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is an ongoing global health emergency [237,238]. SARS-CoV-2 causes an ongoing outbreak of the disease in the lower respiratory tract. SARS-CoV-2 is a highly transmissible and pathogenic coronavirus that emerged in late 2019 and has caused a pandemic of acute respiratory disease, named ‘coronavirus disease 2019′ (COVID-19), which threatens human health and public safety. As of 15 June 2021, there have been 175,987,176 confirmed cases of COVID-19, and 3,811,561 deaths have been reported worldwide. It has affected at least 210 countries. Chan et al. [239]. reported evidence of human-to-human transmission of SARS-CoV-2. SARS-CoV-2 shares 79% genome sequence identity with SARS-CoV and 50% with MERS-CoV [237]. SARS-CoV-2 has six functional open reading frames (ORFs), which are listed in order from 5′ to 3′: replicase (ORF1a/ORF1b), spike (S), envelope (E), membrane (M), and nucleocapsid (N). SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) for infection [240]. SARS-CoV-2 infects cells through proteolytic processing of the S protein, which activates the entry of SARS-CoV-2, and involves transmembrane protease serine protease 2 (TMPRSS2), cathepsin L, and furin [241,242,243]. The pathogenesis of SARS-CoV-2 begins when it binds to epithelial cells in the respiratory tract. After that, SARS-CoV-2 starts replicating and migrates down into the airways, where it enters alveolar epithelial cells in the lungs, initiates replication, and eventually triggers a strong immune response. The replication of SARS-CoV-2 can induce cytokine storm syndrome, which causes acute respiratory distress syndrome and respiratory failure, ultimately resulting in death [13,244,245,246,247,248]. Recently, it has been found that COVID-19 infection results in a weakened immune system, which makes it harder for the body to effectively prevent infection. As a result, COVID-19 patients are at risk for mucormycosis, which is a rare form of infection.
Mucormycosis is an uncommon invasive fungal infection. It is a destructive, life-threatening infection that occurs in people with a susceptible immune system, including people with uncontrolled diabetes mellitus, people who have low levels of neutrophils, or those who are undergoing immunosuppression as part of their treatment for blood cancer, hematopoietic stem cell transplantation, or solid-organ transplantation [249]. The most common symptom is a sinus infection (sinusitis) that is accompanied by nasal congestion, nasal discharge, and sinus pain. Sometimes, blurry vision or double vision can develop, followed by vision loss. Examples of mucormycosis-causing fungi include Rhizopus spp., Mucor spp., Rhizomucor spp., Syncephalastrum spp., Cunninghamella bertholletiae, Apophysomyces spp., and Lichtheimia spp. One study that involved Zataria multiflora essential oil (ZEO) and Zataria multiflora essential oil-loaded solid lipid nanoparticles (ZE-SLNs) with an average size of 255.5 ± 3 nm showed that the ZEO and ZE-SLNs inhibited the growth of Aspergillus ochraceus, A. niger, A. flavus, Alternaria solani, Rhizoctonia solani, and Rhizopus stolonifera by 54 and 79%, respectively [250]. Schiff base ligand 2-(4,6-dimethoxypyrimidin-2-ylimino)methyl)-6-methoxyphenol (DPMM) stabilized copper nanoparticles (DPMM-CuNPs) and nickel nanoparticles exhibited dose-dependent antibacterial and antifungal effects. Studies of the activity of DPMM-CuNPs and DPMM-NiNPs against Shigella sonnei, S. aureus, E. coli, K. pneumoniae, Pseudomonas fluoroscens, and A. niger, C. albicans, C. tropicalis, Mucor indicus and Rhizopus spp. [251]. ZnONPs significantly inhibited the growth of three postharvest fungal isolates including A. alternata, Rhizopus stolonifer, and Botrytis cinerea in a dose-dependent manner [252] Saussurea lappa plant root mediated ZnO NPs with an average size of 123.5 nm also showed significant antifungal activity against A. niger, A. flavus, F. oxysporum, and Rhizopus oryzae [253]. The spherical shape of AgNPs synthesized using the aqueous extract of red seaweed (Gelidiella acerosa) showed antifungal effects against Humicola insolens (MTCC 4520), Fusarium dimerum (MTCC 6583), Mucor indicus (MTCC 3318) and Trichoderma reesei (MTCC 3929). Mucor hiemalis-derived AgNPs exhibited activity against six pathological bacterial strains, namely K. pneumoniae, P. brassicacearum, A. hydrophila, E. coli, B. cereus, and S. aureus, and three pathological fungal strains, namely C. albicans, F. oxysporum, and A. flavus. The antimicrobial activity of AgNPs is comparable to that of antibiotics and fungicides [254]. Dilshad et al. [255]. reported that nanosized silver particles (AgNPs) and micro-sized silver particles (AgMPs) exhibited antibacterial activity against gram-positive bacteria, such as S. aureus, Bacillus subtilis, and Micrococcus luteus, and gram-negative bacteria, including Salmonella setubal, Enterobacter aerogenes, and Agrobacterium tumefaciens. Silver nanoparticles demonstrated antifungal activity by inhibiting the growth of five fungal species: Mucorn spp., A. niger, A. flavus, A. fumigatus, and F. solani.

10. Black Fungus Co-Infection in the Context of the Global COVID-19 Outbreak

COVID-19 is linked to a variety of opportunistic bacterial and fungal diseases [256]. Aspergillosis and Candida-mediated infections have been identified as the primary fungal infections associated with co-infection in COVID-19 patients [9]. Recently, studies reported that mucormycosis in COVID-19 patients have been documented globally, most notably in India. Infection with COVID-19 is linked to fungal infection. Mucormycosis is an acute and deadly fungal infection caused by Mucorales-related fungal species. Uncontrolled diabetes mellitus (DM) and other immunosuppressive diseases such as neutropenia and corticosteroid treatment have been identified as risk factors for mucormycosis. Inhalation of spores that develop into angioinvasive hyphae can cause endothelial damage, resulting in local hemorrhage, thrombosis, and necrosis, as well as the eventual spread of the infection to numerous organs [220,257,258]. (Figure 3). Mucormycosis is an invasive fungal infection, often associated with extremely severe complications in immunocompromised patients [67]. COVID-19-associated mucormycosis (CAM) infections include rhinoorbital mucormycosis, pulmonary mucormycosis, and invasive fungal sinusitis. Several pathogenic fungi, such as A. fumigatus, Rhizopus microspores, Lichtheimia ramosa, and R. arrhizus have been associated with COVID-19 patients (Table 2) [259,260,261,262,263,264,265]. For instance, Iran initially found 15 CAM infections, mainly rhino-orbital mucormycosis, during the first wave of cases from April to September 2020 [67,266,267,268,269]. The rate of mortality was significantly increased due to the association between black fungus infection and COVID-19 infection [72]. The prevalence of mucormycosis (approximately 0.14 cases per 1000 population) in India is about 80 times higher than in other developed countries [217]. Overall, India reported at least 40,845 cases of mucormycosis and 3129 fatalities from such fungal agents since April 2021. Of the total number of mucormycosis patients in India, 34,940 had COVID-19, 26,187 had the co-morbidity of diabetes, and 21,523 were being treated with steroids. Substantial mortality was observed in patients with COVID-19 and diabetes ketoacidosis (DKA) [270,271,272]. Recently, a review reported 101 cases of mucormycosis in COVID-19 patients, of which 82 cases were from India. Of these patients, 18 (out of a total of 31) died due to multiple complications [272]. India has the second-highest diabetes burden in the world and has the highest death due to the disease, which may be one of the major reasons for the increasing number of deaths in patients with CAM [273]. Pakistan, Iraq, and Nepal also reported at least 22, 5, and 14 cases of CAM, respectively.
In India, the fatality rate was 50% due to the increasing number of mucormycosis cases in many Indian states and union territories. The main cause of this issue is the overdosage, panic, and injudicious use of corticosteroids among COVID-19 patients, as well as their pre-existing medical history of diabetes. A case study suggested that most black fungus cases were in India, the United States, and Egypt. This corresponded to 73, 10, and 6% of the 99 patients, respectively. The most prevalent comorbidity was diabetes mellitus (85%). The use of glucocorticoids for the management of COVID-19 was observed in 85% of the cases. Rhino-orbital mucormycosis was most common (42%), followed by rhino-orbito-cerebral mucormycosis (24%). Pulmonary mucormycosis was observed in 10 patients (10%) [274]. Generally, black fungus infections predominantly appeared in patients whose health was compromised by the use of steroids and whose immune system was significantly weakened. An increase in cases of mucormycosis in patients with COVID-19 is prevalent, primarily due to the increased use of steroids, such as dexamethasone, especially among patients with diabetes.

11. Management and Guidelines to Control COVID-19 Lead Black Fungus Infection

“Prevention is better than cure;” hence, measures are necessary to prevent COVID-19 from causing black fungal infections. First, early diagnosis via histopathology or direct microscopic examination and culture, along with molecular methods, such as sequencing common DNA region of fungi known as internal transcribed spacer (ITS) region, is essential [217]. A second important step is the proper management and treatment of patients with CAM, including the proper removal of the infected hard and soft tissue by surgery, as well as parenteral antifungal therapy with Amphotericin B.5 or antifungal drugs such as Posaconazole, Voriconazole and Itraconazole [71]. The third step is to ensure the proper and limited usage of steroid drugs for COVID-19 patients. Critical analysis is essential for the administration of drugs for fungal coinfections. The fourth step is the use of nonpharmacologic measures, such as masking policies and social distancing, which should be taken to reduce the risk of transmission of SARS-CoV-2. Vaccination is also necessary to reduce the spread of COVID-19. International efforts are needed to replenish the critical healthcare supplies and the materials required for manufacturing vaccines. The fifth step is to develop strategies to decrease the risk of mucormycosis in individuals infected with SARS-CoV-2. The sixth step is to ensure, people should avoid environments in which exposure to Mucorales is likely, and to educate healthcare providers and the public about the signs and symptoms of mucormycosis. Hospitals should also have adequate supplies of amphotericin B. It will also be necessary to make strengthening infrastructure and improving healthcare delivery systems high priorities to forestall such crises.
To restrict COVID-19 lead black fungus infections, the following criteria are essential: controlling diabetes and diabetic ketoacidosis, reducing the use of steroids, and regulating the use of immunomodulating drugs. To decrease the number of COVID-19-related black fungus infections, early screening, and testing protocols are imperative. The implementation of patient education is important because it is the best tool to prevent an increase in life-threatening mucormycotic infections. Government efforts should focus on training an ample workforce and on generating technical expertise. Epidemiological surveys must consider that COVID-19 infections can lead to black fungus infection. Other important aspects include preventing the creation of damp areas that could be a breeding ground for mucormycosis and ensuring that rapid diagnosis and treatment methods are widely available.to prevent the impact of this fungal infection. Amphotericin B is one of the best choices for the treatment of mucormycosis. Liposomal amphotericin B with a low dose of 5 mg/day to a higher dose of 10 mg/kg/day for cerebral infection [275,276]. Clinical improvement was observed with the combination of amphotericin B with triazoles such as posaconazole, isavuconazole, voriconazole, etc [277]. Caspofungin exhibited inhibitory activity against Mucorales, and a combination of amphotericin B and caspofungin showed potential synergistic effects against Mucorales. Caspofungin is effective in inhibiting the (1,3)-β-D-glucan synthetase enzyme expressed by R. oryzae [278]. In addition to chemotherapy, adjunctive therapies can be promoted to control mucormycosis infection, such as the administration of iron chelators and the use of hyperbaric oxygen to suppress the growth of mucormycosis-causing fungi, as higher oxygen pressures improve the ability of neutrophils to kill the fungi [279,280].

12. Conclusions and Future Perspectives

COVID-19, caused by SARS-CoV-2, has the potential to become a long-lasting global health crisis. The use of certain steroids in patients with COVID-19 compromises their immunity, and consequently, secondary infections can develop. Patients with COVID-19 have been administered a variety of steroid drugs that weaken their immune systems, and the long-term use of steroids has been found to be associated with mucormycosis or aspergillosis. Recently, the rapidly and continually developing field of nanotechnology has been widely applied in biomedical research. Nanotechnology is a viable prospective treatment, and nanomaterials based on metal ions exhibited strong efficacy against bacteria, fungi, and viruses. Typical antimicrobial agents should have a broad spectrum, high efficiency, and long-lasting antimicrobial activity. They should also be stable and have excellent biocompatibility. The use of nanoparticles as antifungal agents is possible because of their unique antimicrobial properties. As antimicrobial agents, nanoparticles have a broad antimicrobial spectrum and can inhibit the growth of fungi. In this review, we have discussed the impact of silver, zinc oxide, gold, copper, titanium, magnetic, iron, and carbon nanoparticles on various types of fungi. In general, nanoparticles inhibit fungal cell viability by allowing the binding of Ag+ to sulfhydryl (thiol) and phosphate groups in the cell wall of fungi, which leads to interference with the electron transport chain and the generation of energy. Furthermore, nanoparticles inhibit DNA replication and the respiratory chain in fungi, ultimately causing cell death by the generation of ROS. Although nanoparticles have significant applications in biomedicine, concrete guidelines should be devised by experts from industries, governments, and the scientific community to reduce black fungal infection by using nanoparticles. Nanoparticles are promising tools for immune modulation, either by stimulating or suppressing the immune response.
To control COVID-19-related black fungus infection, rapid diagnosis and intervention are essential. This includes controlling blood sugar levels, the urgent removal of dead tissue, and treatment with antifungal drugs. Controlling these fungal infections will require increased awareness and better tests to diagnose them early, along with a focus on controlling diabetes and using corticosteroids appropriately. Patients require access to timely surgery and antifungal treatment. However, further research is also required to learn more about how to prevent such infections. Because of its rapid progression, angioinvasive nature, the swift diagnosis of mucormycosis is essential, and treatment should be initiated as quickly as possible after diagnosis. Early diagnosis, tight glycemic control, and elimination of steroid use are recommended. Prompt radical surgical debridement, liposomal amphotericin B, and posaconazole are the current standard of care. The role of dietary nutraceuticals can be evaluated as a supplemental measure. To overcome the rapid increase of infections caused by the delta variant, quicker diagnoses, better treatment methods, and more research are needed [281].
CAM is an emerging global threat and a significant challenge during the COVID-19 pandemic. CAM is governed by environmental, host, and iatrogenic factors. The involvement of all these factors needs to be evaluated to determine the reason for the unprecedented surge in CAM cases globally. The molecular mechanism between uncontrolled diabetes and CAM needs to be studied extensively. It will also be necessary to determine the pathogenic potential of Mucorales due to SARS-CoV-2 infection.
Nanoparticles are playing a significant role in extensive antifungal effects; however, several factors need to be considered for biomedical applications including harmful effects, bioavailability, cellular interactions, bio-distribution, and biodegradation level of NPs. The accumulation of NPs in the environment and human body and uptake by biological systems leads to catastrophic consequences such as DNA and membrane damage, protein misfolding, and mitochondrial damage. Hence, clear investigations are necessary for widening their applications and successful commercialization. Further studies are required to understand the development of the unique properties of these mystic particles.

Author Contributions

Conceptualization and writing original draft preparation, S.G.; literature collection, A.R.L.; supervision, validation, project administration and funding acquisition, J.H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by “Basic Science Research Program (2021R1C1C100954611) of Korea National Research Foundation grant”.

Acknowledgments

We thank Ramakrishnan Muthuswamy, Nanjing Forestry University, China for helping with image generation using BioRender software.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gupta, S.; Sahoo, S. Pandemic and mental health of the front-line healthcare workers: A review and implications in the Indian context amidst COVID-19. Gen. Psychiatry 2020, 33, e100284. [Google Scholar] [CrossRef]
  2. Baldin, C.; Ibrahim, A.S. Molecular mechanisms of mucormycosis—The bitter and the sweet. PLoS Pathog. 2017, 13, e1006408. [Google Scholar] [CrossRef] [Green Version]
  3. Petrikkos, G.; Skiada, A.; Lortholary, O.; Roilides, E.; Walsh, T.J.; Kontoyiannis, D.P. Epidemiology and Clinical Manifestations of Mucormycosis. Clin. Infect. Dis. 2012, 54, S23–S34. [Google Scholar] [CrossRef]
  4. Gangneux, J.-P.; Bougnoux, M.-E.; Dannaoui, E.; Cornet, M.; Zahar, J. Invasive fungal diseases during COVID-19: We should be prepared. J. De Mycol. Med. 2020, 30, 100971. [Google Scholar] [CrossRef]
  5. Garcia-Vidal, C.; Sanjuan, G.; Moreno-García, E.; Puerta-Alcalde, P.; Garcia-Pouton, N.; Chumbita, M.; Fernandez-Pittol, M.; Pitart, C.; Inciarte, A.; Bodro, M.; et al. Incidence of co-infections and superinfections in hospitalized patients with COVID-19: A retrospective cohort study. Clin. Microbiol. Infect. 2020, 27, 83–88. [Google Scholar] [CrossRef]
  6. Hoenigl, M. Fungal translocation: A driving force behind the occurrence of non-AIDS events. Clin. Infect. Dis. 2020, 70, 242–244. [Google Scholar] [CrossRef]
  7. Koehler, P.; Cornely, O.A.; Bottiger, B.W.; Dusse, F.; Eichenauer, D.A.; Fuchs, F.; Hallek, M.; Jung, N.; Klein, F.; Persigehl, T.; et al. COVID-19 associated pulmonary aspergillosis. Mycoses 2020, 63, 528–534. [Google Scholar] [CrossRef]
  8. Lansbury, L.; Lim, B.; Baskaran, V.; Lim, W.S. Co-infections in people with COVID-19: A systematic review and meta-analysis. J. Infect. 2020, 81, 266–275. [Google Scholar] [CrossRef]
  9. Song, G.; Liang, G.; Liu, W. Fungal Co-infections Associated with Global COVID-19 Pandemic: A Clinical and Diagnostic Perspective from China. Mycopathologia 2020, 185, 599–606. [Google Scholar] [CrossRef]
  10. Dubey, S.; Mukherjee, D.; Sarkar, P.; Mukhopadhyay, P.; Barman, D.; Bandopadhyay, M.; Pandit, A.; Sengupta, A.; Das, S.; Ghosh, S.; et al. COVID-19 associated rhino-orbital-cerebral mucormycosis: An observational study from Eastern India, with special emphasis on neurological spectrum. Diabetes Metab. Syndr. Clin. Res. Rev. 2021, 15, 102267. [Google Scholar] [CrossRef]
  11. Mekki, S.O.; Hassan, A.A.; Falemban, A.; Alkotani, N.; Alsharif, S.M.; Haron, A.; Felemban, B.; Iqbal, M.S.; Tabassum, A. Pulmonary mucormycosis: A case report of a rare infection with potential diagnostic problems. Case Rep. Pathol. 2020, 2020, 4. [Google Scholar] [CrossRef]
  12. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
  13. Huang, J.; He, Y.; Su, Q.; Yang, J. Characteristics of COVID-19 clinical trials in China based on the registration data on ChiCTR and ClinicalTrials.gov. Drug Des. Dev. Ther. 2020, 14, 2159. [Google Scholar] [CrossRef]
  14. Ruan, P.-S.; Xu, H.-Q.; Wu, J.-H.; Song, Q.-F.; Qiu, H.-Y. COVID-19 in Children: Clinical Characteristics and Follow-Up Study. SN Compr. Clin. Med. 2020, 2, 1713–1716. [Google Scholar] [CrossRef]
  15. Lescure, F.-X.; Bouadma, L.; Nguyen, D.; Parisey, M.; Wicky, P.-H.; Behillil, S.; Gaymard, A.; Bouscambert-Duchamp, M.; Donati, F.; Le Hingrat, Q. Clinical and virological data of the first cases of COVID-19 in Europe: A case series. Lancet Infect. Dis. 2020, 20, 697–706. [Google Scholar] [CrossRef] [Green Version]
  16. Van Arkel, A.L.; Rijpstra, T.A.; Belderbos, H.N.; Van Wijngaarden, P.; Verweij, P.E.; Bentvelsen, R.G. COVID-19–associated pulmonary aspergillosis. Am. J. Respir. Crit. Care Med. 2020, 202, 132–135. [Google Scholar] [CrossRef]
  17. Soliman, S.S.M.; Baldin, C.; Gu, Y.; Singh, S.; Gebremariam, T.; Swidergall, M.; Alqarihi, A.; Youssef, E.G.; Alkhazraji, S.; Pikoulas, A.; et al. Mucoricin is a ricin-like toxin that is critical for the pathogenesis of mucormycosis. Nat. Microbiol. 2021, 6, 313–326. [Google Scholar] [CrossRef]
  18. Shaikh, S.; Nazam, N.; Rizvi, S.M.D.; Ahmad, K.; Baig, M.H.; Lee, E.J.; Choi, I. Mechanistic Insights into the Antimicrobial Actions of Metallic Nanoparticles and Their Implications for Multidrug Resistance. Int. J. Mol. Sci. 2019, 20, 2468. [Google Scholar] [CrossRef] [Green Version]
  19. Pereira, L.; Dias, N.; Carvalho, J.; Fernandes, S.; Santos, C.; Lima, N. Synthesis, characterization and antifungal activity of chemically and fungal-produced silver nanoparticles against Trichophyton rubrum. J. Appl. Microbiol. 2014, 117, 1601–1613. [Google Scholar] [CrossRef] [Green Version]
  20. Sardi, J.; Scorzoni, L.; Bernardi, T.; Fusco-Almeida, A.; Giannini, M.M. Candida species: Current epidemiology, pathogenicity, biofilm formation, natural antifungal products and new therapeutic options. J. Med. Microbiol. 2013, 62, 10–24. [Google Scholar] [CrossRef]
  21. Su, S.-Y.; Chao, C.-M. Invasive Candida Infections in Critically Ill Patients. Crit. Care Med. 2015, 43, e322–e323. [Google Scholar] [CrossRef] [PubMed]
  22. Shen, J.; Cowen, L.E.; Griffin, A.M.; Chan, L.; Köhler, J.R. The Candida albicans pescadillo homolog is required for normal hypha-to-yeast morphogenesis and yeast proliferation. Proc. Natl. Acad. Sci. USA 2008, 105, 20918–20923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Bilici, S.; Durna, Y.M.; Yigit, O.; Karatas, A.; Cimen, C.; Fincanci, M.; Gökduman, A.R. The Effect of Mupirocin- and Fusidic Acid-nasal Packings, Placed after Septoplasty, on the Nasal Bacterial Profile. Allergy Rhinol. 2016, 7, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Niemirowicz, K.; Piktel, E.; Wilczewska, A.Z.; Markiewicz, K.H.; Durnaś, B.; Wątek, M.; Puszkarz, I.; Wróblewska, M.; Niklińska, W.; Savage, P.B.; et al. Core–shell magnetic nanoparticles display synergistic antibacterial effects against Pseudomonas aeruginosa and Staphylococcus aureus when combined with cathelicidin LL-37 or selected ceragenins. Int. J. Nanomed. 2016, 11, 5443–5455. [Google Scholar] [CrossRef] [Green Version]
  25. Voltan, A.R.; Quindos, G.; Alarcón, K.P.M.; Fusco-Almeida, A.M.; Mendes-Giannini, M.J.S.; Chorilli, M. Fungal diseases: Could nanostructured drug delivery systems be a novel paradigm for therapy? Int. J. Nanomed. 2016, 11, 3715. [Google Scholar] [CrossRef] [Green Version]
  26. Brandelli, A. Nanostructures as Promising Tools for Delivery of Antimicrobial Peptides. Mini Rev. Med. Chem. 2012, 12, 731–741. [Google Scholar] [CrossRef]
  27. Ananias, H.; De Jong, I.; Dierckx, R.; De Wiele, C.V.; Helfrich, W.; Elsinga, P. Nuclear imaging of prostate cancer with gastrin-releasing-peptide-receptor targeted radiopharmaceuticals. Curr. Pharm. Des. 2008, 14, 3033–3047. [Google Scholar] [CrossRef]
  28. Zhang, C.; Chen, M.; Wang, G.; Fang, W.; Ye, C.; Hu, H.; Fa, Z.; Yi, J.; Liao, W.-Q. Pd@Ag Nanosheets in Combination with Amphotericin B Exert a Potent Anti-Cryptococcal Fungicidal Effect. PLoS ONE 2016, 11, e0157000. [Google Scholar] [CrossRef] [Green Version]
  29. Dizaj, S.M.; Vazifehasl, Z.; Salatin, S.; Adibkia, K.; Javadzadeh, Y. Nanosizing of drugs: Effect on dissolution rate. Res. Pharm. Sci. 2015, 10, 95. [Google Scholar]
  30. Hajipour, M.J.; Fromm, K.M.; Ashkarran, A.A.; de Aberasturi, D.J.; de Larramendi, I.R.; Rojo, T.; Serpooshan, V.; Parak, W.J.; Mahmoudi, M. Antibacterial properties of nanoparticles. Trends Biotechnol. 2012, 30, 499–511. [Google Scholar] [CrossRef] [Green Version]
  31. Mal’tseva, V.N.; Goltyaev, M.V.; Turovsky, E.A.; Varlamova, E.G. Immunomodulatory and Anti-Inflammatory Properties of Selenium-Containing Agents: Their Role in the Regulation of Defense Mechanisms against COVID-19. Int. J. Mol. Sci. 2022, 23, 2360. [Google Scholar] [CrossRef]
  32. Gurunathan, S.; Han, J.W.; Kwon, D.-N.; Kim, J.-H. Enhanced antibacterial and anti-biofilm activities of silver nanoparticles against Gram-negative and Gram-positive bacteria. Nanoscale Res. Lett. 2014, 9, 373. [Google Scholar] [CrossRef] [Green Version]
  33. Gurunathan, S.; Jeong, J.-K.; Han, J.W.; Zhang, X.-F.; Park, J.H.; Kim, J.-H. Multidimensional effects of biologically synthesized silver nanoparticles in Helicobacter pylori, Helicobacter felis, and human lung (L132) and lung carcinoma A549 cells. Nanoscale Res. Lett. 2015, 10, 35. [Google Scholar] [CrossRef] [Green Version]
  34. Wani, I.A.; Ahmad, T. Size and shape dependant antifungal activity of gold nanoparticles: A case study of Candida. Colloids Surf. B Biointerfaces 2013, 101, 162–170. [Google Scholar] [CrossRef]
  35. Birla, S.S.; Gaikwad, S.C.; Gade, A.K.; Rai, M.K. Rapid synthesis of silver nanoparticles from Fusarium oxysporum by optimizing physicocultural conditions. Sci. World J. 2013, 2013, 12. [Google Scholar] [CrossRef] [Green Version]
  36. Robinson J, R.; Isikhuemhen, O.S.; Anike, F.N. Fungal-Metal Interactions: A Review of Toxicity and Homeostasis. J. Fungi 2021, 7, 225. [Google Scholar] [CrossRef]
  37. Gudikandula, K.; Vadapally, P.; Charya, M.S. Biogenic synthesis of silver nanoparticles from white rot fungi: Their characterization and antibacterial studies. OpenNano 2017, 2, 64–78. [Google Scholar] [CrossRef]
  38. Jamdagni, P.; Rana, J.; Khatri, P. Comparative study of antifungal effect of green and chemically synthesised silver nanoparticles in combination with carbendazim, mancozeb, and thiram. IET Nanobiotechnol. 2018, 12, 1102–1107. [Google Scholar] [CrossRef]
  39. Varshney, R.; Bhadauria, S.; Gaur, M.S. A Review: Biological Synthesis of Silver and Copper Nanoparticles. Nano Biomed. Eng. 2012, 4, 99–106. [Google Scholar] [CrossRef] [Green Version]
  40. Peters, V.; Tumkur, T.; Ma, J.; Kotov, N.A.; Noginov, M. Strong coupling of localized surface plasmons and ensembles of dye molecules. Optics Express 2016, 24, 25653–25664. [Google Scholar] [CrossRef]
  41. Franci, G.; Falanga, A.; Galdiero, S.; Palomba, L.; Rai, M.; Morelli, G.; Galdiero, M. Silver Nanoparticles as Potential Antibacterial Agents. Molecules 2015, 20, 8856–8874. [Google Scholar] [CrossRef]
  42. Lemire, J.A.; Harrison, J.J.; Turner, R.J. Antimicrobial activity of metals: Mechanisms, molecular targets and applications. Nat. Rev. Microbiol. 2013, 11, 371–384. [Google Scholar] [CrossRef]
  43. Gurunathan, S.; Han, J.W.; Eppakayala, V.; Jeyaraj, M.; Kim, J.-H. Cytotoxicity of Biologically Synthesized Silver Nanoparticles in MDA-MB-231 Human Breast Cancer Cells. Bio. Med. Res. Int. 2013, 2013, 535796. [Google Scholar] [CrossRef] [Green Version]
  44. Yang, Y.; Gao, N.; Hu, Y.; Jia, C.; Chou, T.; Du, H.; Wang, H. Gold nanoparticle-enhanced photodynamic therapy: Effects of surface charge and mitochondrial targeting. Ther. Deliv. 2015, 6, 307–321. [Google Scholar] [CrossRef]
  45. Zhao, Y.; Zhang, W.; Lin, Y.; Du, D. The vital function of Fe3O4@Au nanocomposites for hydrolase biosensor design and its application in detection of methyl parathion. Nanoscale 2013, 5, 1121–1126. [Google Scholar] [CrossRef]
  46. De Alteriis, E.; Maselli, V.; Falanga, A.; Galdiero, S.; Di Lella, F.M.; Gesuele, R.; Guida, M.; Galdiero, E. Efficiency of gold nanoparticles coated with the antimicrobial peptide indolicidin against biofilm formation and development of Candida spp. clinical isolates. Infect. Drug Resist. 2018, 11, 915. [Google Scholar] [CrossRef] [Green Version]
  47. Becheri, A.; Dürr, M.; Lo Nostro, P.; Baglioni, P. Synthesis and characterization of zinc oxide nanoparticles: Application to textiles as UV-absorbers. J. Nanoparticle Res. 2008, 10, 679–689. [Google Scholar] [CrossRef]
  48. Rasmussen, J.W.; Martinez, E.; Louka, P.; Wingett, D.G. Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications. Expert Opin. Drug Deliv. 2010, 7, 1063–1077. [Google Scholar] [CrossRef] [Green Version]
  49. Bai, K.-J.; Chuang, K.-J.; Ma, C.-M.; Chang, T.-Y.; Chuang, H.-C. Human lung adenocarcinoma cells with an EGFR mutation are sensitive to non-autophagic cell death induced by zinc oxide and aluminium-doped zinc oxide nanoparticles. J. Toxicol. Sci. 2017, 42, 437–444. [Google Scholar] [CrossRef] [Green Version]
  50. Ravi, K.S.; Ranjana, G. Synthesis of zinc oxide nanoparticles by homogeneous precipitation method and its application in antifungal activity against Candida albicans. Ceram. Int. 2015, 41, 967–975. [Google Scholar]
  51. Gurunathan, S.; Han, J.W.; Dayem, A.A.; Eppakayala, V.; Kim, J.-H. Oxidative stress-mediated antibacterial activity of graphene oxide and reduced graphene oxide in Pseudomonas aeruginosa. Int. J. Nanomed. 2012, 7, 5901–5914. [Google Scholar] [CrossRef] [Green Version]
  52. Kang, S.; Pinault, M.; Pfefferle, L.D.; Elimelech, M. Single-Walled Carbon Nanotubes Exhibit Strong Antimicrobial Activity. Langmuir 2007, 23, 8670–8673. [Google Scholar] [CrossRef]
  53. Manke, A.; Wang, L.; Rojanasakul, Y. Pulmonary toxicity and fibrogenic response of carbon nanotubes. Toxicol. Mech. Methods 2013, 23, 196–206. [Google Scholar] [CrossRef] [Green Version]
  54. Shvedova, A.A.; Pietroiusti, A.; Fadeel, B.; Kagan, V.E. Mechanisms of carbon nanotube-induced toxicity: Focus on oxidative stress. Toxicol. Appl. Pharmacol. 2012, 261, 121–133. [Google Scholar] [CrossRef] [Green Version]
  55. Vecitis, C.D.; Zodrow, K.R.; Kang, S.; Elimelech, M. Electronic-structure-dependent bacterial cytotoxicity of single-walled carbon nanotubes. ACS Nano. 2010, 4, 5471–5479. [Google Scholar] [CrossRef]
  56. YadavHMY, H.M.; Thorat, N.D.; Yallapu, M.M.; Tofail, S.A.; Kim, J.-S. Functional TiO2 nanocoral architecture for light-activated cancer chemotherapy. J. Mater. Chem. 2017, 5, 1461–1470. [Google Scholar]
  57. Zhao, S.; Wang, S.; Yan, J. Experimental study of hydroxyapatite (HA) granules filled around the titanium implant. Zhonghua Kou Qiang Yi Xue Za Zhi Zhonghua Kouqiang Yixue Zazhi = Chin. J. Stomatol. 1998, 33, 353–354. [Google Scholar]
  58. Diebold, U. Structure and properties of TiO2 surfaces: A brief review. Appl. Phys. A 2003, 76, 681–687. [Google Scholar] [CrossRef]
  59. Kumar, R.; Sahoo, G.; Chawla-Sarkar, M.; Nayak, M.; Trivedi, K.; Rana, S.; Pandey, K.; Das, V.; Topno, R.; Das, P. Antiviral effect of Glycine coated Iron oxide nanoparticles iron against H1N1 influenza A virus. Int. J. Infect. Dis. 2016, 45, 281–282. [Google Scholar] [CrossRef] [Green Version]
  60. Shukla, S.; Arora, V.; Jadaun, A.; Kumar, J.; Singh, N.; Jain, V.K. Magnetic removal of Entamoeba cysts from water using chitosan oligosaccharide-coated iron oxide nanoparticles. Int. J. Nanomed. 2015, 10, 4901–4917. [Google Scholar] [CrossRef] [Green Version]
  61. Taylor, E.N.; Webster, T.J. The use of superparamagnetic nanoparticles for prosthetic biofilm prevention. Int. J. Nanomed. 2009, 4, 145–152. [Google Scholar]
  62. Tran, N.; Mir, A.; Mallik, D.; Sinha, A.; Nayar, S.; Webster, T.J. Bactericidal effect of iron oxide nanoparticles on Staphylococcus aureus. Int. J. Nanomed. 2010, 5, 277–283. [Google Scholar] [CrossRef] [Green Version]
  63. Prucek, R.; Tuček, J.; Kilianová, M.; Panáček, A.; Kvítek, L.; Filip, J.; Kolář, M.; Tománková, K.; Zbořil, R. The targeted antibacterial and antifungal properties of magnetic nanocomposite of iron oxide and silver nanoparticles. Biomaterials 2011, 32, 4704–4713. [Google Scholar] [CrossRef]
  64. Tocco, M.P.; Ballardini, M.; Masala, M.; Perozzi, A. Post-sternotomy chronic osteomyelitis: Is sternal resection always necessary? Eur. J. Cardio-Thorac. Surg. 2013, 43, 715–721. [Google Scholar] [CrossRef]
  65. Barnes, R.J.; Riba, O.; Gardner, M.N.; Scott, T.B.; Jackman, S.A.; Thompson, I.P. Optimization of nano-scale nickel/iron particles for the reduction of high concentration chlorinated aliphatic hydrocarbon solutions. Chemosphere 2010, 79, 448–454. [Google Scholar] [CrossRef]
  66. Seabra, A.B.; Haddad, P.; Duran, N. Biogenic synthesis of nanostructured iron compounds: Applications and perspectives. IET Nanobiotechnol. 2013, 7, 90–99. [Google Scholar] [CrossRef]
  67. Chowdhary, A.; Sharma, A. The lurking scourge of multidrug resistant Candida auris in times of COVID-19 pandemic. J. Glob. Antimicrob. Resist. 2020, 22, 175–176. [Google Scholar] [CrossRef]
  68. Pasero, D.; Sanna, S.; Liperi, C.; Piredda, D.; Branca, G.P.; Casadio, L.; Simeo, R.; Buselli, A.; Rizzo, D.; Bussu, F. A challenging complication following SARS-CoV-2 infection: A case of pulmonary mucormycosis. Infection 2021, 49, 1055–1060. [Google Scholar] [CrossRef]
  69. Waizel-Haiat, S.; Guerrero-Paz, J.A.; Sanchez-Hurtado, L.; Calleja-Alarcon, S.; Romero-Gutierrez, L. A Case of Fatal Rhino-Orbital Mucormycosis Associated with New Onset Diabetic Ketoacidosis and COVID-19. Cureus 2021, 13. [Google Scholar] [CrossRef]
  70. Narayanan, S.; Chua, J.V.; Baddley, J.W. Coronavirus disease 2019–associated mucormycosis: Risk factors and mechanisms of disease. Clin. Infect. Dis. 2022, 74, 1279–1283. [Google Scholar] [CrossRef]
  71. Muthu, V.; Rudramurthy, S.M.; Chakrabarti, A.; Agarwal, R. Epidemiology and Pathophysiology of COVID-19-Associated Mucormycosis: India Versus the Rest of the World. Mycopathologia 2021, 186, 739–754. [Google Scholar] [CrossRef]
  72. Garg, D.; Muthu, V.; Sehgal, I.S.; Ramachandran, R.; Kaur, H.; Bhalla, A.; Puri, G.D.; Chakrabarti, A.; Agarwal, R. Coronavirus Disease (COVID-19) Associated Mucormycosis (CAM): Case Report and Systematic Review of Literature. Mycopathologia 2021, 186, 289–298. [Google Scholar] [CrossRef]
  73. Buil, J.B.; van Zanten, A.R.; Bentvelsen, R.G.; Rijpstra, T.A.; Goorhuis, B.; van der Voort, S.; Wammes, L.J.; Janson, J.A.; Melchers, M.; Heusinkveld, M. Case series of four secondary mucormycosis infections in COVID-19 patients, the Netherlands, December 2020 to May 2021. Eurosurveillance 2021, 26, 2100510. [Google Scholar] [CrossRef]
  74. Mukherjee, B.; Raichura, N.D.; Alam, M.S. Fungal infections of the orbit. Indian J. Ophthalmol. 2016, 64, 337. [Google Scholar] [CrossRef]
  75. Millon, L.; Larosa, F.; Lepiller, Q.; Legrand, F.; Rocchi, S.; Daguindau, E.; Scherer, E.; Bellanger, A.-P.; Leroy, J.; Grenouillet, F. Quantitative Polymerase Chain Reaction Detection of Circulating DNA in Serum for Early Diagnosis of Mucormycosis in Immunocompromised Patients. Clin. Infect. Dis. 2013, 56, e95–e101. [Google Scholar] [CrossRef]
  76. Millon, L.; Herbrecht, R.; Grenouillet, F.; Morio, F.; Alanio, A.; Letscher-Bru, V.; Cassaing, S.; Chouaki, T.; Kauffmann-Lacroix, C.; Poirier, P.; et al. Early diagnosis and monitoring of mucormycosis by detection of circulating DNA in serum: Retrospective analysis of 44 cases collected through the French Surveillance Network of Invasive Fungal Infections (RESSIF). Clin. Microbiol. Infect. 2016, 22, 810.e1–810.e8. [Google Scholar] [CrossRef] [Green Version]
  77. Kim, J.S.; Kuk, E.; Yu, K.N.; Kim, J.-H.; Park, S.J.; Lee, H.J.; Kim, S.H.; Park, Y.K.; Park, Y.H.; Hwang, C.-Y. Antimicrobial effects of silver nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2007, 3, 95–101. [Google Scholar] [CrossRef]
  78. Martinez-Gutierrez, F.; Olive, P.L.; Banuelos, A.; Orrantia, E.; Niño, N.; Sanchez, E.M.; Ruiz, F.; Bach, H.; Av-Gay, Y. Synthesis, characterization, and evaluation of antimicrobial and cytotoxic effect of silver and titanium nanoparticles. Nanomedicine 2010, 6, 681–688. [Google Scholar] [CrossRef]
  79. Rai, M.; Yadav, A.; Gade, A. Silver nanoparticles as a new generation of antimicrobials. Biotechnol. Adv. 2009, 27, 76–83. [Google Scholar] [CrossRef]
  80. McShan, D.; Ray, P.C.; Yu, H. Molecular toxicity mechanism of nanosilver. J. Food Drug Anal. 2014, 22, 116–127. [Google Scholar] [CrossRef] [Green Version]
  81. Riaz, M.; Altaf, M.; Faisal, A.; Shekheli, M.A.; Miana, G.A.; Khan, M.Q.; Shah, M.A.; Ilyas, S.Z.; Khan, A.A. Biogenic Synthesis of AgNPs with Saussurea lappa C.B. Clarke and Studies on Their Biochemical Properties. J. Nanosci. Nanotechnol. 2018, 18, 8392–8398. [Google Scholar] [CrossRef]
  82. Radhakrishnan, V.S.; Dwivedi, S.P.; Siddiqui, M.H.; Prasad, T. In vitro studies on oxidative stress-independent, Ag nanoparticles-induced cell toxicity of Candida albicans, an opportunistic pathogen. Int. J. Nanomed. 2018, 13, 91–96. [Google Scholar] [CrossRef] [Green Version]
  83. Alghuthaymi, M.A.; Abd-Elsalam, K.A.; Shami, A.; Said-Galive, E.; Shtykova, E.V.; Naumkin, A.V. Silver/Chitosan nanocomposites: Preparation and characterization and their fungicidal activity against dairy cattle toxicosis Penicillium expansum. J. Fungi 2020, 6, 51. [Google Scholar] [CrossRef] [Green Version]
  84. Das, B.; Khan, M.; Jayabalan, R.; Behera, S.K.; Yun, S.-I.; Tripathy, S.K.; Mishra, A. Understanding the antifungal mechanism of Ag@ ZnO core-shell nanocomposites against Candida krusei. Sci. Rep. 2016, 6, 36403. [Google Scholar] [CrossRef] [Green Version]
  85. Auyeung, A.; Casillas-Santana, M.A.; Martinez-Castanon, G.A.; Slavin, Y.N.; Zhao, W.; Asnis, J.; Häfeli, U.O.; Bach, H. Effective control of molds using a combination of nanoparticles. PLoS ONE 2017, 12, e0169940. [Google Scholar] [CrossRef]
  86. Kim, K.-J.; Sung, W.-S.; Moon, S.-K.; Choi, J.-S.; Kim, J.-G.; Lee, D.-G. Antifungal effect of silver nanoparticles on dermatophytes. J. Microbiol. Biotechnol. 2008, 18, 1482–1484. [Google Scholar]
  87. Kim, J.; Kwon, S.; Ostler, E. Antimicrobial effect of silver-impregnated cellulose: Potential for antimicrobial therapy. J. Biol. Eng. 2009, 3, 20. [Google Scholar] [CrossRef] [Green Version]
  88. Nasrollahi, A.; Pourshamsian, K.; Mansourkiaee, P. Antifungal activity of silver nanoparticles on some of fungi. Int. J. Nanodimens. 2011, 1, 233–239. [Google Scholar]
  89. Gutiérrez, J.A.; Caballero, S.; Díaz, L.A.; Guerrero, M.A.; Ruiz, J.; Ortiz, C.C. High antifungal activity against candida species of monometallic and bimetallic nanoparticles synthesized in nanoreactors. ACS Biomater. Sci. Eng. 2018, 4, 647–653. [Google Scholar] [CrossRef]
  90. He, M.; Lu, L.; Zhang, J.; Li, D. Immobilized Silver Nanoparticles on Chitosan with Special Surface State-Enhanced Antimicrobial Efficacy and Reduced Cytotoxicity. J. Nanosci. Nanotechnol. 2015, 15, 6435–6443. [Google Scholar] [CrossRef]
  91. Pinto, R.J.; Almeida, A.; Fernandes, S.C.; Freire, C.S.; Silvestre, A.J.; Neto, C.P.; Trindade, T. Antifungal activity of transparent nanocomposite thin films of pullulan and silver against Aspergillus niger. Colloids Surf. B Biointerfaces 2013, 103, 143–148. [Google Scholar] [CrossRef]
  92. Panáček, A.; Kolář, M.; Večeřová, R.; Prucek, R.; Soukupová, J.; Kryštof, V.; Hamal, P.; Zbořil, R.; Kvítek, L. Antifungal activity of silver nanoparticles against Candida spp. Biomaterials 2009, 30, 6333–6340. [Google Scholar] [CrossRef]
  93. Mallmann, E.J.; Cunha, F.A.; Castro, B.N.; Maciel, A.M.; Menezes, E.A.; Fechine, P.B. Antifungal activity of silver nanoparticles obtained by green synthesis. Rev. Inst. Med. Trop Sao Paulo 2015, 57, 165–167. [Google Scholar] [CrossRef]
  94. Ogar, A.; Tylko, G.; Turnau, K. Antifungal properties of silver nanoparticles against indoor mould growth. Sci. Total Environ. 2015, 521–522, 305–314. [Google Scholar] [CrossRef]
  95. Xu, F.; Piett, C.; Farkas, S.; Qazzaz, M.; Syed, N.I. Silver nanoparticles (AgNPs) cause degeneration of cytoskeleton and disrupt synaptic machinery of cultured cortical neurons. Mol. Brain 2013, 6, 29. [Google Scholar] [CrossRef] [Green Version]
  96. Qasim, M.; Singh, B.R.; Naqvi, A.; Paik, P.; Das, D. Silver nanoparticles embedded mesoporous SiO2 nanosphere: An effective anticandidal agent against Candida albicans 077. Nanotechnology 2015, 26, 285102. [Google Scholar] [CrossRef]
  97. Bajpai, R.; Yusuf, M.A.; Upreti, D.K.; Gupta, V.K.; Singh, B.N. Endolichenic fungus, Aspergillus quandricinctus of Usnea longissima inhibits quorum sensing and biofilm formation of Pseudomonas aeruginosa PAO1. Microb. Pathog. 2020, 140, 103933. [Google Scholar]
  98. Wunnoo, S.; Paosen, S.; Lethongkam, S.; Sukkurd, R.; Waen-ngoen, T.; Nuidate, T.; Phengmak, M.; Voravuthikunchai, S.P. Biologically rapid synthesized silver nanoparticles from aqueous Eucalyptus camaldulensis leaf extract: Effects on hyphal growth, hydrolytic enzymes, and biofilm formation in Candida albicans. Biotechnol. Bioeng. 2021, 118, 1578–1592. [Google Scholar] [CrossRef]
  99. El Sayed, M.T.; El-Sayed, A.S. Biocidal activity of metal nanoparticles synthesized by Fusarium solani against multidrug-resistant bacteria and mycotoxigenic fungi. J. Microbiol. Biotechnol. 2020, 30, 226–236. [Google Scholar] [CrossRef]
  100. Palmquist, D.L.; Lock, A.L.; Shingfield, K.J.; Bauman, D.E. Biosynthesis of conjugated linoleic acid in ruminants and humans. Adv. Food Nutr. Res. 2005, 50, 179–217. [Google Scholar]
  101. Lipovsky, A.; Nitzan, Y.; Gedanken, A.; Lubart, R. Antifungal activity of ZnO nanoparticles—The role of ROS mediated cell injury. Nanotechnology 2011, 22, 105101. [Google Scholar] [CrossRef]
  102. Gondal, M.A.; Alzahrani, A.J.; Randhawa, M.A.; Siddiqui, M.N. Morphology and antifungal effect of nano-ZnO and nano-Pd-doped nano-ZnO against Aspergillus and Candida. J. Environ. Sci. Health Part A 2012, 47, 1413–1418. [Google Scholar] [CrossRef]
  103. He, L.; Liu, Y.; Mustapha, A.; Lin, M. Antifungal activity of zinc oxide nanoparticles against Botrytis cinerea and Penicillium expansum. Microbiol. Res. 2011, 166, 207–215. [Google Scholar] [CrossRef]
  104. Jalal, M.; Ansari, M.A.; Ali, S.G.; Khan, H.M.; Rehman, S. Anticandidal activity of bioinspired ZnO NPs: Effect on growth, cell morphology and key virulence attributes of Candida species. Artif. Cells Nanomed. Biotechnol. 2018, 46, 912–925. [Google Scholar] [CrossRef] [Green Version]
  105. Khan, S.A.; Shahid, S.; Mahmood, T.; Lee, C.-S. Contact lenses coated with hybrid multifunctional ternary nanocoatings (Phytomolecule-coated ZnO nanoparticles: Gallic Acid: Tobramycin) for the treatment of bacterial and fungal keratitis. Acta Biomater. 2021, 128, 262–276. [Google Scholar] [CrossRef]
  106. Sierra-Fernandez, A.; De la Rosa-Garcia, S.C.; Gomez-Villalba, L.S.; Gómez-Cornelio, S.; Rabanal, M.E.; Fort, R.; Quintana, P. Synthesis, Photocatalytic, and Antifungal Properties of MgO, ZnO and Zn/Mg Oxide Nanoparticles for the Protection of Calcareous Stone Heritage. ACS Appl. Mater. Interfaces 2017, 9, 24873–24886. [Google Scholar] [CrossRef]
  107. Jasim, D.A.; Ménard-Moyon, C.; Bégin, D.; Bianco, A.; Kostarelos, K. Tissue distribution and urinary excretion of intravenously administered chemically functionalized graphene oxide sheets. Chem. Sci. 2015, 6, 3952–3964. [Google Scholar] [CrossRef] [Green Version]
  108. Cierech, M.; Wojnarowicz, J.; Szmigiel, D.; Bączkowski, B.; Grudniak, A.M.; Wolska, K.I.; Łojkowski, W.; Mierzwińska-Nastalska, E. Preparation and characterization of ZnO-PMMA resin nanocomposites for denture bases. Acta Bioeng. Biomech. 2016, 18, 31–41. [Google Scholar]
  109. Lakshmeesha, T.R.; Kalagatur, N.K.; Mudili, V.; Mohan, C.D.; Rangappa, S.; Prasad, B.D.; Ashwini, B.S.; Hashem, A.; Alqarawi, A.A.; Malik, J.A. Biofabrication of zinc oxide nanoparticles with Syzygium aromaticum flower buds extract and finding its novel application in controlling the growth and mycotoxins of Fusarium graminearum. Front. Microbiol. 2019, 10, 1244. [Google Scholar] [CrossRef] [Green Version]
  110. Alkilany, A.M.; Thompson, L.B.; Boulos, S.P.; Sisco, P.N.; Murphy, C.J. Gold nanorods: Their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Adv. Drug Deliv. Rev. 2012, 64, 190–199. [Google Scholar] [CrossRef]
  111. Conde, J.; Bao, C.; Cui, D.; Baptista, P.V.; Tian, F. Antibody–drug gold nanoantennas with Raman spectroscopic fingerprints for in vivo tumour theranostics. J. Control. Release 2014, 183, 87–93. [Google Scholar] [CrossRef] [Green Version]
  112. Wang, Q.; Tang, H.; Xie, Q.; Jia, X.; Zhang, Y.; Tan, L.; Yao, S. The preparation and characterization of poly (o-phenylenediamine)/gold nanoparticles interface for immunoassay by surface plasmon resonance and electrochemistry. Colloids Surf. B Biointerfaces 2008, 63, 254–261. [Google Scholar] [CrossRef]
  113. Brown, S.D.; Nativo, P.; Smith, J.-A.; Stirling, D.; Edwards, P.R.; Venugopal, B.; Flint, D.J.; Plumb, J.A.; Graham, D.; Wheate, N.J. Gold nanoparticles for the improved anticancer drug delivery of the active component of oxaliplatin. J. Am. Chem. Soc. 2010, 132, 4678–4684. [Google Scholar] [CrossRef]
  114. Huang, Y.-F.; Chang, H.-T.; Tan, W. Cancer cell targeting using multiple aptamers conjugated on nanorods. Anal. Chem. 2008, 80, 567–572. [Google Scholar] [CrossRef]
  115. Hernández-Sierra, J.F.; Ruiz, F.; Pena, D.C.C.; Martínez-Gutiérrez, F.; Martínez, A.E.; Guillén, A.d.J.P.; Tapia-Pérez, H.; Castañón, G.M. The antimicrobial sensitivity of Streptococcus mutans to nanoparticles of silver, zinc oxide, and gold. Nanomed. Nanotechnol. Biol. Med. 2008, 4, 237–240. [Google Scholar] [CrossRef]
  116. Lima, E.; Guerra, R.; Lara, V.; Guzmán, A. Gold nanoparticles as efficient antimicrobial agents for Escherichia coli and Salmonella typhi. Chem. Central J. 2013, 7, 11. [Google Scholar] [CrossRef] [Green Version]
  117. Khan, A.U.; Khan, S.; Azam, A.; Alam, F. Gold nanoparticles enhance methylene blue–induced photodynamic therapy: A novel therapeutic approach to inhibit Candida albicans biofilm. Int. J. Nanomed. 2012, 7, 3245–3257. [Google Scholar] [CrossRef] [Green Version]
  118. Jebali, A.; Hajjar, F.H.E.; Pourdanesh, F.; Hekmatimoghaddam, S.; Kazemi, B.; Masoudi, A.; Daliri, K.; Sedighi, N. Silver and gold nanostructures: Antifungal property of different shapes of these nanostructures on Candida species. Med. Mycol. 2014, 52, 65–72. [Google Scholar]
  119. Seong, M.; Lee, D.G. Reactive oxygen species-independent apoptotic pathway by gold nanoparticles in Candida albicans. Microbiol. Res. 2018, 207, 33–40. [Google Scholar] [CrossRef]
  120. Yu, Q.; Li, J.; Zhang, Y.; Wang, Y.; Liu, L.; Li, M. Inhibition of gold nanoparticles (AuNPs) on pathogenic biofilm formation and invasion to host cells. Sci. Rep. 2016, 6, 26667. [Google Scholar] [CrossRef]
  121. Bajaj, M.; Pandey, S.K.; Nain, T.; Brar, S.K.; Singh, P.; Singh, S.; Wangoo, N.; Sharma, R.K. Stabilized cationic dipeptide capped gold/silver nanohybrids: Towards enhanced antibacterial and antifungal efficacy. Colloids Surf. B Biointerf. 2017, 158, 397–407. [Google Scholar] [CrossRef]
  122. Khatoon, N.; Alam, H.; Manzoor, N.; Sardar, M. Removal of toxic contaminants from water by sustainable green synthesised non-toxic silver nanoparticles. IET Nanobiotechnol. 2018, 12, 1090–1096. [Google Scholar] [CrossRef]
  123. Milanezi, F.G.; Meireles, L.M.; de Christo Scherer, M.M.; de Oliveira, J.P.; da Silva, A.R.; de Araujo, M.L.; Endringer, D.C.; Fronza, M.; Guimarães, M.C.C.; Scherer, R. Antioxidant, antimicrobial and cytotoxic activities of gold nanoparticles capped with quercetin. Saudi Pharm. J. 2019, 27, 968–974. [Google Scholar] [CrossRef]
  124. Rodriguez-Torres, M.D.P.; Díaz-Torres, L.A.; Millán-Chiu, B.E.; García-Contreras, R.; Hernández-Padrón, G.; Acosta-Torres, L.S. Antifungal and cytotoxic evaluation of photochemically synthesized heparin-coated gold and silver nanoparticles. Molecules 2020, 25, 2849. [Google Scholar] [CrossRef]
  125. Rahimi, H.; Roudbarmohammadi, S.; H, H.D.; Roudbary, M. Antifungal effects of indolicidin-conjugated gold nanoparticles against fluconazole-resistant strains of Candida albicans isolated from patients with burn infection. Int. J. Nanomed. 2019, 14, 5323–5338. [Google Scholar] [CrossRef] [Green Version]
  126. Chalandar, H.E.; Ghorbani, H.R.; Attar, H.; Alavi, S.A. Antifungal effect of copper and copper oxide nanoparticles against Penicillium on orange fruit. Biosci. Biotechnol. Res. Asia 2017, 14, 279–284. [Google Scholar] [CrossRef]
  127. Shende, S.; Ingle, A.P.; Gade, A.; Rai, M. Green synthesis of copper nanoparticles by Citrus medica Linn. (Idilimbu) juice and its antimicrobial activity. World, J. Microbiol. Biotechnol. 2015, 31, 865–873. [Google Scholar] [CrossRef]
  128. Usman, M.S.; El Zowalaty, M.E.; Shameli, K.; Zainuddin, N.; Salama, M.; Ibrahim, N.A. Synthesis, characterization, and antimicrobial properties of copper nanoparticles. Int. J. Nanomed. 2013, 8, 4467–4479. [Google Scholar] [CrossRef] [Green Version]
  129. Oussou-Azo, A.F.; Nakama, T.; Nakamura, M.; Futagami, T.; Vestergaard, M.D.C.M. Antifungal potential of nanostructured crystalline copper and its oxide forms. Nanomaterials 2020, 10, 1003. [Google Scholar] [CrossRef]
  130. Arendsen, L.P.; Vig, S.; Thakar, R.; Sultan, A.H. Impact of copper compression stockings on venous insufficiency and lipodermatosclerosis: A randomised controlled trial. Phlebology 2019, 34, 224–230. [Google Scholar] [CrossRef]
  131. Grass, G.; Rensing, C.; Solioz, M. Metallic copper as an antimicrobial surface. Appl. Environ. Microbiol. 2011, 77, 1541–1547. [Google Scholar] [CrossRef] [PubMed]
  132. Weaver, L.; Noyce, J.O.; Michels, H.T.; Keevil, C.W. Potential action of copper surfaces on meticillin-resistant Staphylococcus aureus. J. Appl. Microbiol. 2010, 109, 2200–2205. [Google Scholar] [CrossRef] [PubMed]
  133. Mani, V.M.; Kalaivani, S.; Sabarathinam, S.; Vasuki, M.; Soundari, A.J.P.G.; Das, M.A.; Elfasakhany, A.; Pugazhendhi, A. Copper oxide nanoparticles synthesized from an endophytic fungus Aspergillus terreus: Bioactivity and anti-cancer evaluations. Environ. Res. 2021, 201, 111502. [Google Scholar] [CrossRef] [PubMed]
  134. Eremenko, A.M.; Petrik, I.S.; Smirnova, N.P.; Rudenko, A.V.; Marikvas, Y.S. Antibacterial and antimycotic activity of cotton fabrics, impregnated with silver and binary silver/copper nanoparticles. Nanoscale Res. Lett. 2016, 11, 28. [Google Scholar] [CrossRef] [Green Version]
  135. Viet, P.V.; Nguyen, H.T.; Cao, T.M.; Hieu, L.V. Fusarium antifungal activities of copper nanoparticles synthesized by a chemical reduction method. J. Nanomater. 2016, 2016, 1957612. [Google Scholar] [CrossRef] [Green Version]
  136. Pariona, N.; Mtz-Enriquez, A.I.; Sánchez-Rangel, D.; Carrión, G.; Paraguay-Delgado, F.; Rosas-Saito, G. Green-synthesized copper nanoparticles as a potential antifungal against plant pathogens. RSC Adv. 2019, 9, 18835–18843. [Google Scholar] [CrossRef] [Green Version]
  137. Devipriya, D.; Roopan, S.M. Cissus quadrangularis mediated ecofriendly synthesis of copper oxide nanoparticles and its antifungal studies against Aspergillus niger, Aspergillus flavus. Mater. Sci Eng C Mater. Biol. Appl. 2017, 80, 38–44. [Google Scholar] [CrossRef]
  138. Roopan, S.M.; Priya, D.D.; Shanavas, S.; Acevedo, R.; Al-Dhabi, N.A.; Arasu, M.V. CuO/C nanocomposite: Synthesis and optimization using sucrose as carbon source and its antifungal activity. Mater. Sci. Eng. C 2019, 101, 404–414. [Google Scholar] [CrossRef]
  139. Ammar, H.Y.; Badran, H.M. Effect of CO adsorption on properties of transition metal doped porphyrin: A DFT and TD-DFT study. Heliyon 2019, 5, e2545. [Google Scholar] [CrossRef] [Green Version]
  140. Asghar, M.A.; Zahir, E.; Shahid, S.M.; Khan, M.N.; Asghar, M.A.; Iqbal, J.; Walker, G. Iron, copper and silver nanoparticles: Green synthesis using green and black tea leaves extracts and evaluation of antibacterial, antifungal and aflatoxin B1 adsorption activity. LWT 2018, 90, 98–107. [Google Scholar] [CrossRef] [Green Version]
  141. Henam, S.D.; Ahmad, F.; Shah, M.A.; Parveen, S.; Wani, A.H. Microwave synthesis of nanoparticles and their antifungal activities. Spectrochim. Acta Part. A Mol. Biomol. Spectrosc. 2019, 213, 337–341. [Google Scholar] [CrossRef]
  142. Hermida-Montero, L.A.; Pariona, N.; Mtz-Enriquez, A.I.; Carrión, G.; Paraguay-Delgado, F.; Rosas-Saito, G. Aqueous-phase synthesis of nanoparticles of copper/copper oxides and their antifungal effect against Fusarium oxysporum. J. Hazard. Mater. 2019, 380, 120850. [Google Scholar] [CrossRef]
  143. Padmavathi, A.R.; Das, A.; Priya, A.; Sushmitha, T.J.; Pandian, S.K.; Toleti, S.R. Impediment to growth and yeast-to-hyphae transition in Candida albicans by copper oxide nanoparticles. Biofouling 2020, 36, 56–72. [Google Scholar] [CrossRef]
  144. Martínez, A.; Apip, C.; Meléndrez, M.; Domínguez, M.; Sánchez-Sanhueza, G.; Marzialetti, T.; Catalán, A. Dual antifungal activity against Candida albicans of copper metallic nanostructures and hierarchical copper oxide marigold-like nanostructures grown in situ in the culture medium. J. Appl. Microbiol. 2020, 130, 1883–1892. [Google Scholar] [CrossRef]
  145. Akturk, A.; Güler, F.K.; Taygun, M.E.; Goller, G.; Küçükbayrak, S. Synthesis and antifungal activity of soluble starch and sodium alginate capped copper nanoparticles. Mater. Res. Express 2020, 6, 1250g3. [Google Scholar] [CrossRef]
  146. Consolo, V.F.; Torres-Nicolini, A.; Alvarez, V.A. Mycosinthetized Ag, CuO and ZnO nanoparticles from a promising Trichoderma harzianum strain and their antifungal potential against important phytopathogens. Sci. Rep. 2020, 10, 20499. [Google Scholar] [CrossRef]
  147. Rajakumar, G.; Rahuman, A.A.; Roopan, S.M.; Khanna, V.G.; Elango, G.; Kamaraj, C.; Zahir, A.A.; Velayutham, K. Fungus-mediated biosynthesis and characterization of TiO(2) nanoparticles and their activity against pathogenic bacteria. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2012, 91, 23–29. [Google Scholar] [CrossRef]
  148. Ahmad, M.A.; Yuesuo, Y.; Ao, Q.; Adeel, M.; Hui, Z.Y.; Javed, R. Appraisal of Comparative Therapeutic Potential of Undoped and Nitrogen-Doped Titanium Dioxide Nanoparticles. Molecules 2019, 24, 3916. [Google Scholar] [CrossRef] [Green Version]
  149. Zacarias, S.M.; Marchetti, S.; Alfano, O.M.; de los Milagros Ballari, M. Photocatalytic paint for fungi growth control under different environmental conditions and irradiation sources. J. Photochem. Photobiol. A Chem. 2018, 364, 364. [Google Scholar] [CrossRef]
  150. Zuccheri, T.; Colonna, M.; Stefanini, I.; Santini, C.; Di Gioia, D. Bactericidal Activity of Aqueous Acrylic Paint Dispersion for Wooden Substrates Based on TiO2 Nanoparticles Activated by Fluorescent Light. Materials 2013, 6, 3270–3283. [Google Scholar] [CrossRef]
  151. Moradpoor, H.; Safaei, M.; Golshah, A.; Mozaffari, H.R.; Sharifi, R.; Imani, M.M.; Mobarakeh, M.S. Green synthesis and antifungal effect of titanium dioxide nanoparticles on oral Candida albicans pathogen. Inorg. Chem. Commun. 2021, 130, 108748. [Google Scholar] [CrossRef]
  152. Mukherjee, M.; Gangopadhyay, K.; Das, R.; Purkayastha, P. Development of Non-ionic Surfactant and Protein-Coated Ultrasmall Silver Nanoparticles: Increased Viscoelasticity Enables Potency in Biological Applications. ACS Omega 2020, 5, 8999–9006. [Google Scholar] [CrossRef] [Green Version]
  153. Kermani, F.; Sadeghian, M.; Shokohi, T.; Hashemi, S.; Moslemi, D.; Davodian, S.; Abastabar, M.; Bandalizadeh, Z.; Faeli, L.; Seifi, Z.; et al. Molecular identification and antifungal susceptibility testing of Candida species isolated from oral lesions in patients with head and neck cancer undergoing radiotherapy. Curr. Med. Mycol. 2021, 7, 44–50. [Google Scholar] [CrossRef]
  154. Ilkhechi, N.N.; Mozammel, M.; Khosroushahi, A.Y. Antifungal effects of ZnO, TiO2 and ZnO-TiO2 nanostructures on Aspergillus flavus. Pestic. Biochem. Physiol. 2021, 176, 104869. [Google Scholar] [CrossRef]
  155. Maness, P.C.; Smolinski, S.; Blake, D.M.; Huang, Z.; Wolfrum, E.J.; Jacoby, W.A. Bactericidal activity of photocatalytic TiO(2) reaction: Toward an understanding of its killing mechanism. Appl. Environ. Microbiol. 1999, 65, 4094–4098. [Google Scholar] [CrossRef] [Green Version]
  156. Singh, P. Biosynthesis of titanium dioxide nanoparticles and their antibacterial property. Int. J. Chem. Mol. Eng. 2016, 10, 275–278. [Google Scholar]
  157. Carré, G.; Hamon, E.; Ennahar, S.; Estner, M.; Lett, M.-C.; Horvatovich, P.; Gies, J.-P.; Keller, V.; Keller, N.; Andre, P. TiO2 Photocatalysis Damages Lipids and Proteins in Escherichia coli. Appl. Environ. Microbiol. 2014, 80, 2573–2581. [Google Scholar] [CrossRef] [Green Version]
  158. Kubacka, A.; Suarez-Diez, M.; Rojo, D.; Bargiela, R.; Ciordia, S.; Zapico, I.; Albar, J.P.; Barbas, C.; Dos Santos, V.A.P.M.; Fernández-García, M.; et al. Understanding the antimicrobial mechanism of TiO2-based nanocomposite films in a pathogenic bacterium. Sci. Rep. 2014, 4, 4134. [Google Scholar] [CrossRef] [Green Version]
  159. Korshed, P.; Li, L.; Liu, Z.; Mironov, A.; Wang, T. Antibacterial mechanisms of a novel type picosecond laser-generated silver-titanium nanoparticles and their toxicity to human cells. Int. J. Nanomed. 2017, 13, 89–101. [Google Scholar] [CrossRef] [Green Version]
  160. Chawengkijwanich, C.; Hayata, Y. Development of TiO2 powder-coated food packaging film and its ability to inactivate Escherichia coli in vitro and in actual tests. Int. J. Food Microbiol. 2008, 123, 288–292. [Google Scholar] [CrossRef]
  161. Maneerat, C.; Hayata, Y. Antifungal activity of TiO2 photocatalysis against Penicillium expansum in vitro and in fruit tests. Int. J. Food Microbiol. 2006, 107, 99–103. [Google Scholar] [CrossRef]
  162. Muzzarelli, R.A. Human enzymatic activities related to the therapeutic administration of chitin derivatives. Cell Mol. Life Sci. 1997, 53, 131–140. [Google Scholar] [CrossRef]
  163. Jackson, S.L.; Heath, I.B. Roles of calcium ions in hyphal tip growth. Microbiol. Rev. 1993, 2, 367–382. [Google Scholar] [CrossRef]
  164. Pham, T.D.; Lee, B.K. Effects of Ag doping on the photocatalytic disinfection of E. coli in bioaerosol by Ag-TiO(2)/GF under visible light. J. Colloid Interface Sci. 2014, 428, 24–31. [Google Scholar] [CrossRef]
  165. Nithya, A.; JeevaKumari, H.L.; Rokesh, K.; Ruckmani, K.; Jeganathan, K.; Jothivenkatachalam, K. A versatile effect of chitosan-silver nanocomposite for surface plasmonic photocatalytic and antibacterial activity. J. Photochem. Photobiol. B Biol. 2015, 153, 412–422. [Google Scholar] [CrossRef]
  166. Ashkarran, A.A.; Hamidinezhad, H.; Haddadi, H.; Mahmoudi, M. Double-doped TiO2 nanoparticles as an efficient visible-light-active photocatalyst and antibacterial agent under solar simulated light. Appl. Surf. Sci. 2014, 301, 338–345. [Google Scholar] [CrossRef]
  167. Kong, M.; Chen, X.G.; Xing, K.; Park, H.J. Antimicrobial properties of chitosan and mode of action: A state of the art review. Int. J. Food Microbiol. 2010, 144, 51–63. [Google Scholar] [CrossRef]
  168. Fujishima, A.; Zhang, X.; Tryk, D.A. TiO2 photocatalysis and related surface phenomena. Surf. Sci. Rep. 2008, 63, 515–582. [Google Scholar] [CrossRef]
  169. Wei, X.; Yu, J.; Ding, L.; Hu, J.; Jiang, W. Effect of oxide nanoparticles on the morphology and fluidity of phospholipid membranes and the role of hydrogen bonds. J. Environ. Sci. 2017, 57, 221–230. [Google Scholar] [CrossRef]
  170. Reddy, S.; Porter, D.; Patton, J.T.; Al-Nafussi, A.; Beggs, I. Endometriosis of the superior gluteal nerve. Skelet. Radiol. 2007, 36, 879–883. [Google Scholar] [CrossRef]
  171. Nehra, P.; Chauhan, R.; Garg, N.; Verma, K. Antibacterial and antifungal activity of chitosan coated iron oxide nanoparticles. Br. J. Biomed. Sci. 2017, 75, 13–18. [Google Scholar] [CrossRef]
  172. Alam, T.; Khan, R.A.A.; Ali, A.; Sher, H.; Ullah, Z.; Ali, M. Biogenic synthesis of iron oxide nanoparticles via Skimmia laureola and their antibacterial efficacy against bacterial wilt pathogen Ralstonia solanacearum. Mater. Sci. Eng. C 2018, 98, 101–108. [Google Scholar] [CrossRef]
  173. Seddighi, N.S.; Salari, S.; Izadi, A.R. Evaluation of antifungal effect of iron-oxide nanoparticles against different Candida species. IET Nanobiotechnol. 2017, 11, 883–888. [Google Scholar] [CrossRef]
  174. Niemirowicz, K.; Durnaś, B.; Tokajuk, G.; Głuszek, K.; Wilczewska, A.; Misztalewska-Turkowicz, I.; Mystkowska, J.; Michalak, G.; Sodo, A.; Wątek, M.; et al. Magnetic nanoparticles as a drug delivery system that enhance fungicidal activity of polyene antibiotics. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 2395–2404. [Google Scholar] [CrossRef]
  175. Niemirowicz, K.; Durnaś, B.; Tokajuk, G.; Piktel, E.; Michalak, G.; Gu, X.; Kułakowska, A.; Savage, P.B.; Bucki, R. Formulation and candidacidal activity of magnetic nanoparticles coated with cathelicidin LL-37 and ceragenin CSA-13. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
  176. Lara, H.H.; Romero-Urbina, D.G.; Pierce, C.; Lopez-Ribot, J.L.; Arellano-Jiménez, M.J.; Jose-Yacaman, M. Effect of silver nanoparticles on Candida albicans biofilms: An ultrastructural study. J. Nanobiotechnol. 2015, 13, 91. [Google Scholar] [CrossRef] [Green Version]
  177. Hasany, S.F.; Abdurahman, N.H.; Sunarti, A.R.; Jose, R. Magnetic Iron Oxide Nanoparticles: Chemical Synthesis and Applications Review. Curr. Nanosci. 2013, 9, 561–575. [Google Scholar] [CrossRef]
  178. Abbas, H.S.; Krishnan, A. Magnetic Nanosystems as a Therapeutic Tool to Combat Pathogenic Fungi. Adv. Pharm. Bull. 2020, 10, 512–523. [Google Scholar] [CrossRef]
  179. Devi, H.S.; Boda, M.A.; Shah, M.A.; Parveen, S.; Wani, A.H. Green synthesis of iron oxide nanoparticles using Platanus orientalis leaf extract for antifungal activity. Green Process. Synth. 2019, 8, 38–45. [Google Scholar] [CrossRef]
  180. De Lima, T.M.; Arias, L.S.; Afanaci, L.F.; Ferraresse, R.F.; Neto, F.N.D.S.; De Lima, B.H.; Straioto, F.G.; De Camargo, E.R.; Pessan, J.P.; Monteiro, D.R. Assembly and antifungal effect of a new fluconazole-carrier nanosystem. Future Microbiol. 2020, 15, 273–285. [Google Scholar] [CrossRef]
  181. Li, C.; Wang, X.; Chen, F.; Zhang, C.; Zhi, X.; Wang, K.; Cui, D. The antifungal activity of graphene oxide–silver nanocomposites. Biomaterials 2013, 34, 3882–3890. [Google Scholar] [CrossRef] [PubMed]
  182. Patel, M.B.; Harikrishnan, U.; Valand, N.N.; Modi, N.R.; Menon, S.K. Novel Cationic Quinazolin-4 (3H)-one Conjugated Fullerene Nanoparticles as Antimycobacterial and Antimicrobial Agents. Arch. Der Pharm. 2013, 346, 210–220. [Google Scholar] [CrossRef] [PubMed]
  183. Diez-Orejas, R.; Feito, M.J.; Cicuéndez, M.; Casarrubios, L.; Rojo, J.M.; Portolés, M.T. Graphene oxide nanosheets increase Candida albicans killing by pro-inflammatory and reparative peritoneal macrophages. Colloids Surf. B Biointerfaces 2018, 171, 250–259. [Google Scholar] [CrossRef] [PubMed]
  184. Li, X.; Huang, R.; Tang, F.K.; Li, W.C.; Wong, S.S.W.; Leung, K.C.F.; Jin, L. Red-Emissive Guanylated Polyene-Functionalized Carbon Dots Arm Oral Epithelia against Invasive Fungal Infections. ACS Appl. Mater. Interfaces 2019, 11, 46591–46603. [Google Scholar] [CrossRef]
  185. Jhonsi, M.A.; Ananth, D.A.; Nambirajan, G.; Sivasudha, T.; Yamini, R.; Bera, S.; Kathiravan, A. Antimicrobial activity, cytotoxicity and DNA binding studies of carbon dots. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2018, 196, 295–302. [Google Scholar] [CrossRef]
  186. Priyadarshini, E.; Rawat, K.; Prasad, T.; Bohidar, H. Antifungal efficacy of Au@ carbon dots nanoconjugates against opportunistic fungal pathogen, Candida albicans. Colloids Surf. B Biointerfaces 2018, 163, 355–361. [Google Scholar] [CrossRef]
  187. Sawangphruk, M.; Srimuk, P.; Chiochan, P.; Sangsri, T.; Siwayaprahm, P. Synthesis and antifungal activity of reduced graphene oxide nanosheets. Carbon 2012, 50, 5156–5161. [Google Scholar] [CrossRef]
  188. Wang, X.; Liu, X.; Han, H. Evaluation of antibacterial effects of carbon nanomaterials against copper-resistant Ralstonia solanacearum. Colloids Surf. B Biointerfaces 2013, 103, 136–142. [Google Scholar] [CrossRef]
  189. Xie, J.; Ming, Z.; Li, H.; Yang, H.; Yu, B.; Wu, R.; Liu, X.; Bai, Y.; Yang, S.-T. Toxicity of graphene oxide to white rot fungus Phanerochaete chrysosporium. Chemosphere 2016, 151, 324–331. [Google Scholar] [CrossRef]
  190. Akhavan, O.; Ghaderi, E.; Akhavan, A. Size-dependent genotoxicity of graphene nanoplatelets in human stem cells. Biomaterials 2012, 33, 8017–8025. [Google Scholar] [CrossRef]
  191. Miragoli, M.; Novak, P.; Ruenraroengsak, P.; Shevchuk, A.I.; Korchev, Y.E.; Lab, M.J.; Tetley, T.D.; Gorelik, J. Functional interaction between charged nanoparticles and cardiac tissue: A new paradigm for cardiac arrhythmia? Nanomedicine 2013, 8, 725–737. [Google Scholar] [CrossRef] [Green Version]
  192. Savi, M.; Rossi, S.; Bocchi, L.; Gennaccaro, L.; Cacciani, F.; Perotti, A.; Amidani, D.; Alinovi, R.; Goldoni, M.; Aliatis, I.; et al. Titanium dioxide nanoparticles promote arrhythmias via a direct interaction with rat cardiac tissue. Part. Fibre Toxicol. 2014, 11, 63. [Google Scholar] [CrossRef]
  193. Nguyen, D.H.; Vo, T.N.N.; Nguyen, N.T.; Ching, Y.C.; Hoang Thi, T.T. Comparison of biogenic silver nanoparticles formed by Momordica charantia and Psidium guajava leaf extract and antifungal evaluation. PLoS ONE 2020, 15, e0239360. [Google Scholar] [CrossRef]
  194. Jebril, N.M.T. Evaluation of two fixation techniques for direct observation of biofilm formation of Bacillus subtilis in situ, on Congo red agar, using scanning electron microscopy. Vet. World 2020, 13, 1133–1137. [Google Scholar] [CrossRef]
  195. Krishnaraj, C.; Ramachandran, R.; Mohan, K.; Kalaichelvan, P. Optimization for rapid synthesis of silver nanoparticles and its effect on phytopathogenic fungi. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2012, 93, 95–99. [Google Scholar] [CrossRef]
  196. Jafari, A.A.; Tafti, A.F.; Hoseiny, S.M.; Kazemi, A. Antifungal Effect of Zataria multiflora Essence on Experimentally Contaminated Acryl Resin Plates with Candida albicans. Iran. Red Crescent Med. J. 2015, 17, e16552. [Google Scholar] [CrossRef] [Green Version]
  197. Huang, W.; Fang, X.; Wang, H.; Chen, F.; Duan, H.; Bi, Y.; Yu, H. Biosynthesis of AgNPs by B. maydis and its antifungal effect against Exserohilum turcicum. IET Nanobiotechnol. 2018, 12, 585–590. [Google Scholar] [CrossRef]
  198. Sahayaraj, K.; Rajesh, S.; Rathi, J.A.M.; Kumar, V. Green preparation of seaweed-based silver nano-liquid for cotton pathogenic fungi management. IET Nanobiotechnol. 2018, 13, 219–225. [Google Scholar] [CrossRef]
  199. Ediz, C.; Tavukcu, H.H.; Akan, S.; Kizilkan, Y.E.; Alcin, A.; Oz, K.; Yilmaz, O. Is there any association of COVID-19 with testicular pain and epididymo-orchitis? Int. J. Clin. Pract. 2021, 75, e13753. [Google Scholar] [CrossRef]
  200. Soumdararajan, H.C.; Bullock, S.L. The influence of dynein processivity control, MAPs, and microtubule ends on directional movement of a localising mRNA. Elife 2014, 3, e01596. [Google Scholar] [CrossRef]
  201. Al-Otibi, F.; Perveen, K.; Al-Saif, N.A.; Alharbi, R.I.; Bokhari, N.A.; Albasher, G.; Al-Otaibi, R.M.; Al-Mosa, M.A. Biosynthesis of silver nanoparticles using Malva parviflora and their antifungal activity. Saudi J. Biol. Sci. 2021, 28, 2229–2235. [Google Scholar] [CrossRef]
  202. Asghar, F.; Yan, H.; Jiang, L. The putative transcription factor CaMaf1 controls the sensitivity to lithium and rapamycin and represses RNA polymerase III transcription in Candida albicans. FEMS Yeast Res. 2018, 18, foy068. [Google Scholar] [CrossRef]
  203. Abd-El-Aziz, A.S.; Agatemor, C.; Etkin, N.; Overy, D.P.; Lanteigne, M.; McQuillan, K.; Kerr, R.G. Antimicrobial Organometallic Dendrimers with Tunable Activity against Multidrug-Resistant Bacteria. Biomacromolecules 2015, 16, 3694–3703. [Google Scholar] [CrossRef]
  204. Bholay, A.D.; Nalawade, P.M.; Borkhataria, B.V. Fungicidal potential of biosynthesized silver nanoparticles against phytopatho gens and potentiation of fluconazole. World J. Pharm. Res. 2013, 1, 12–15. [Google Scholar]
  205. El-Saadony, M.T.; Saad, A.M.; Najjar, A.A.; Alzahrani, S.O.; Alkhatib, F.M.; Shafi, M.E.; Selem, E.; Desoky, E.S.M.; Fouda, S.E.; El-Tahan, A.M.; et al. The use of biological selenium nanoparticles to suppress Triticum aestivum L. crown and root rot diseases induced by Fusarium species and improve yield under drought and heat stress. Saudi J. Biol Sci. 2021, 28, 4461–4471. [Google Scholar] [CrossRef]
  206. Win, T.T.; Khan, S.; Bo, B.; Zada, S.; Fu, P. Green synthesis and characterization of Fe3O4 nanoparticles using Chlorella-K01 extract for potential enhancement of plant growth stimulating and antifungal activity. Sci. Rep. 2021, 11, 21996. [Google Scholar] [CrossRef]
  207. Fernández, L.; Mediano, P.; García, R.; Rodríguez, J.M.; Marín, M. Risk Factors Predicting Infectious Lactational Mastitis: Decision Tree Approach versus Logistic Regression Analysis. Matern. Child Health J. 2016, 20, 1895–1903. [Google Scholar] [CrossRef]
  208. Rajeshkumar, S.; Rinitha, G. Nanostructural characterization of antimicrobial and antioxidant copper nanoparticles syn thesized using novel Persea americana seeds. OpenNano 2018, 3, 18–27. [Google Scholar] [CrossRef]
  209. Hasanin, M.; Elbahnasawy, M.A.; Shehabeldine, A.M.; Hashem, A.H. Ecofriendly preparation of silver nanoparticles-based nanocomposite stabilized by polysaccharides with antibacterial, antifungal and antiviral activities. Biometals 2021, 34, 1313–1328. [Google Scholar] [CrossRef]
  210. Hassan, M.M.; Ranzoni, A.; Cooper, M.A. A nanoparticle-based method for culture-free bacterial DNA enrichment from whole blood. Biosens. Bioelectron. 2018, 99, 150–155. [Google Scholar] [CrossRef]
  211. Nemati, S.; Hassanzadeh, R.; Jahromi, S.K.; Abadi, A.D.N. Otomycosis in the north of Iran: Common pathogens and resistance to antifungal agents. Eur. Arch. Otorhinolaryngol. 2013, 271, 953–957. [Google Scholar] [CrossRef]
  212. Yassin, M.T.; Mostafa, A.A.F.; Al-Askar, A.A.; Al-Otibi, F.O. Facile Green Synthesis of Zinc Oxide Nanoparticles with Potential Synergistic Activity with Common Antifungal Agents against Multidrug-Resistant Candidal Strains. Crystals 2022, 12, 774. [Google Scholar] [CrossRef]
  213. Kermani, S.A.; Salari, S.; Almani, P.G.N. Comparison of antifungal and cytotoxicity activities of titanium dioxide and zinc oxide nanoparticles with amphotericin B against different Candida species: In vitro evaluation. J. Clin. Lab. Anal. 2021, 35, e23577. [Google Scholar] [CrossRef]
  214. Filipović, N.; Ušjak, D.; Milenković, M.T.; Zheng, K.; Liverani, L.; Boccaccini, A.R.; Stevanović, M.M. Comparative Study of the Antimicrobial Activity of Selenium Nanoparticles with Different Surface Chemistry and Structure. Front. Bioeng. Biotechnol. 2021, 8, 624621. [Google Scholar] [CrossRef]
  215. Jeong, W.; Keighley, C.; Wolfe, R.; Lee, W.L.; Slavin, M.A.; Kong, D.C.M.; Chen, S.C.-A. The epidemiology and clinical manifestations of mucormycosis: A systematic review and meta-analysis of case reports. Clin. Microbiol. Infect. 2019, 25, 26–34. [Google Scholar] [CrossRef] [Green Version]
  216. Rajendra, S.A.; Muddana, K.; Bakki, S.R. Fungal Infections of Oral Cavity: Diagnosis, Management, and Association with COVID-19. SN Compr. Clin. Med. 2021, 3, 1373–1384. [Google Scholar] [CrossRef]
  217. Skiada, A.; Pavleas, I.; Drogari-Apiranthitou, M. Epidemiology and Diagnosis of Mucormycosis: An Update. J. Fungi 2020, 6, 265. [Google Scholar] [CrossRef]
  218. Binder, U.; Maurer, E.; Lass-Florl, C. Mucormycosis—From the pathogens to the disease. Clin. Microbiol Infect. 2014, 20, 60–66. [Google Scholar] [CrossRef] [Green Version]
  219. Ibrahim, A.S. Host cell invasion in mucormycosis: Role of iron. Curr. Opin. Microbiol. 2011, 14, 406–411. [Google Scholar] [CrossRef] [Green Version]
  220. Ahmadikia, K.; Hashemi, S.J.; Khodavaisy, S.; Getso, M.I.; Alijani, N.; Badali, H.; Mirhendi, H.; Salehi, M.; Tabari, A.; Ardehali, M.M.; et al. The double-edged sword of systemic corticosteroid therapy in viral pneumonia: A case report and comparative review of influenza-associated mucormycosis versus COVID-19 associated mucormycosis. Mycoses 2021, 64, 798–808. [Google Scholar] [CrossRef]
  221. Schoen, C.; Reichard, U.; Monod, M.; Kratzin, H.D.; Rüchel, R. Molecular cloning of an extracellular aspartic proteinase from Rhizopus microsporus and evidence for its expression during infection. Med. Mycol. 2002, 40, 61–71. [Google Scholar] [CrossRef] [Green Version]
  222. Spreer, A.; Rüchel, R.; Reichard, U. Characterization of an extracellular subtilisin protease of Rhizopus microsporus and evidence for its expression during invasive rhinoorbital mycosis. Med. Mycol. 2006, 44, 723–731. [Google Scholar] [CrossRef] [Green Version]
  223. Ben-Ami, R.; Luna, M.; Lewis, R.E.; Walsh, T.J.; Kontoyiannis, D.P. A clinicopathological study of pulmonary mucormycosis in cancer patients: Extensive angioinvasion but limited inflammatory response. J. Infect. 2009, 59, 134–138. [Google Scholar] [CrossRef]
  224. Jose, A.; Singh, S.; Roychoudhury, A.; Kholakiya, Y.; Arya, S.; Roychoudhury, S. Current Understanding in the Pathophysiology of SARS-CoV-2-Associated Rhino-Orbito-Cerebral Mucormycosis: A Comprehensive Review. J. Maxillofac. Oral Surg. 2021, 20, 373–380. [Google Scholar] [CrossRef]
  225. Ibrahim, A.S.; Bowman, J.C.; Avanessian, V.; Brown, K.; Spellberg, B.; Edwards, J.E., Jr.; Douglas, C.M. Caspofungin inhibits Rhizopus oryzae 1,3-beta-D-glucan synthase, lowers burden in brain measured by quantitative PCR, and improves survival at a low but not a high dose during murine disseminated zygomycosis. Antimicrob. Agents Chemother. 2005, 49, 721–727. [Google Scholar] [CrossRef] [Green Version]
  226. Alqarihi, A.; Gebremariam, T.; Gu, Y.; Swidergall, M.; Alkhazraji, S.; Soliman, S.S.M.; Bruno, V.M.; Edwards, J.E.; Filler, S.G.; Uppuluri, P.; et al. GRP78 and Integrins Play Different Roles in Host Cell Invasion during Mucormycosis. mBio 2020, 11, e01087-20. [Google Scholar] [CrossRef]
  227. Liu, M.; Spellberg, B.; Phan, Q.T.; Fu, Y.; Fu, Y.; Lee, A.; Edwards, J.E.; Filler, S.G.; Ibrahim, A.S. The endothelial cell receptor GRP78 is required for mucormycosis pathogenesis in diabetic mice. J. Clin. Investig. 2010, 120, 1914–1924. [Google Scholar] [CrossRef] [Green Version]
  228. Bhatt, K.; Agolli, A.; Patel, M.H.; Garimella, R.; Devi, M.; Garcia, E.; Amin, H.; Domingue, C.; Del Castillo, R.G.; Sanchez-Gonzalez, M. High mortality co-infections of COVID-19 patients: Mucormycosis and other fungal infections. Discoveries 2021, 9, e126. [Google Scholar] [CrossRef]
  229. Deb, A.K.; Sarkar, S.; Gokhale, T.; Choudhury, S.S. COVID-19 and orbital mucormycosis. Indian J. Ophthalmol. 2021, 69, 1002–1004. [Google Scholar] [CrossRef]
  230. Ibrahim, M.A.; Sany, D.; El Shahawy, Y.; Awdallah, A. Effect of activated vitamin D on glucoparameters in HCV seropositive and seronegative patients on chronic hemodialysis. Ren. Fail. 2012, 34, 1188–1194. [Google Scholar] [CrossRef] [Green Version]
  231. Stanford, F.A.; Voigt, K. Iron Assimilation during Emerging Infections Caused by Opportunistic Fungi with emphasis on Mucorales and the Development of Antifungal Resistance. Genes 2020, 11, 1296. [Google Scholar] [CrossRef] [PubMed]
  232. Lecube, A.; Hernández, C.; Genescà, J.; Esteban, J.I.; Jardí, R.; García, L.; Simó, R. Diabetes Is the Main Factor Accounting for the High Ferritin Levels Detected in Chronic Hepatitis C Virus Infection. Diabetes Care 2004, 27, 2669–2675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Habib, H.M.; Ibrahim, S.; Zaim, A.; Ibrahim, W.H. The role of iron in the pathogenesis of COVID-19 and possible treatment with lactoferrin and other iron chelators. Biomed. Pharmacother. 2021, 136, 111228. [Google Scholar] [CrossRef]
  234. Zhou, C.; Chen, Y.; Ji, Y.; He, X.; Xue, D. Increased Serum Levels of Hepcidin and Ferritin Are Associated with Severity of COVID-19. Med. Sci. Monit. 2020, 26, e926178-1. [Google Scholar] [CrossRef]
  235. Gómez-Pastora, J.; Weigand, M.; Kim, J.; Wu, X.; Strayer, J.; Palmer, A.F.; Zborowski, M.; Yazer, M.; Chalmers, J.J. Hyperferritinemia in critically ill COVID-19 patients—Is ferritin the product of inflammation or a pathogenic mediator? Clin. Chim. Acta 2020, 509, 249–251. [Google Scholar] [CrossRef]
  236. Cui, J.; Li, F.; Shi, Z.-L. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 2019, 17, 181–192. [Google Scholar] [CrossRef] [Green Version]
  237. Deng, L.S.; Yuan, J.; Ding, L.; Chen, Y.L.; Zhao, C.H.; Chen, G.Q.; Li, X.H.; Li, X.H.; Luo, W.T.; Lan, J.F.; et al. Comparison of patients hospitalized with COVID-19, H7N9 and H1N1. Infect. Dis. Poverty 2020, 9, 163. [Google Scholar] [CrossRef]
  238. Han, Y.J.; Ren, Z.G.; Li, X.X.; Yan, J.L.; Ma, C.Y.; Wu, D.D.; Ji, X.Y. Advances and challenges in the prevention and treatment of COVID-19. Int. J. Med. Sci. 2020, 17, 1803–1810. [Google Scholar] [CrossRef]
  239. Chan, K.H.; Sridhar, S.; Zhan, R.R.; Chu, H.; Fung, A.F.; Chan, G.; Chan, J.W.; To, K.W.; Hung, I.N.; Cheng, V.C.; et al. Factors affecting stability and infectivity of SARS-CoV-2. J. Hosp. Infect. 2020, 106, 226–231. [Google Scholar] [CrossRef]
  240. Zhou, Z.; Kang, H.; Li, S.; Zhao, X. Understanding the neurotropic characteristics of SARS-CoV-2: From neurological manifestations of COVID-19 to potential neurotropic mechanisms. J. Neurol. 2020, 267, 2179–2184. [Google Scholar] [CrossRef]
  241. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  242. Ou, R.; Mo, L.; Tang, H.; Leng, S.; Zhu, H.; Zhao, L.; Ren, Y.; Xu, Y. circRNA-AKT1 Sequesters miR-942-5p to Upregulate AKT1 and Promote Cervical Cancer Progression. Mol. Ther. Nucleic Acids 2020, 20, 308–322. [Google Scholar] [CrossRef]
  243. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  244. Liu, Y.; Lv, J.; Liu, J.; Li, M.; Xie, J.; Lv, Q.; Deng, W.; Zhou, N.; Zhou, Y.; Song, J.; et al. Mucus production stimulated by IFN-AhR signaling triggers hypoxia of COVID-19. Cell Res. 2020, 30, 1078–1087. [Google Scholar] [CrossRef]
  245. Tian, Y.; Rong, L.; Nian, W.; He, Y. Review article: Gastrointestinal features in COVID-19 and the possibility of faecal transmission. Aliment. Pharmacol. Ther. 2020, 51, 843–851. [Google Scholar] [CrossRef]
  246. Wu, Y.Y.; Wang, S.H.; Wu, C.H.; Yen, L.C.; Lai, H.F.; Ho, C.L.; Chiu, Y.L. In silico immune infiltration profiling combined with functional enrichment analysis reveals a potential role for naive B cells as a trigger for severe immune responses in the lungs of COVID-19 patients. PLoS ONE 2020, 15, e242900. [Google Scholar] [CrossRef]
  247. Yao, Y.; Wang, H.; Liu, Z. Expression of ACE2 in airways: Implication for COVID-19 risk and disease management in patients with chronic inflammatory respiratory diseases. Clin. Exp. Allergy 2020, 50, 1313–1324. [Google Scholar] [CrossRef]
  248. Mehta, N.S.; Mytton, O.T.; Mullins, E.W.; Fowler, T.A.; Falconer, C.L.; Murphy, O.B.; Langenberg, C.; Jayatunga, W.J.; Eddy, D.H.; Nguyen-Van-Tam, J.S. SARS-CoV-2 (COVID-19): What Do We Know about Children? A Systematic Review. Clin. Infect. Dis. 2020, 71, 2469–2479. [Google Scholar] [CrossRef]
  249. Farmakiotis, D.; Kontoyiannis, D.P. Mucormycoses. Infect Dis. Clin. North Am. 2016, 30, 143–163. [Google Scholar] [CrossRef]
  250. Nasseri, M.; Golmohammadzadeh, S.; Arouiee, H.; Jaafari, M.R.; Neamati, H. Antifungal activity of Zataria multiflora essential oil-loaded solid lipid nanoparticles in-vitro condition. Iran. J. Basic Med. Sci. 2016, 19, 1231. [Google Scholar]
  251. Jose, P.A.; Raja, J.D.; Sankarganesh, M.; Rajesh, J. Evaluation of antioxidant, DNA targeting, antimicrobial and cytotoxic studies of imine capped copper and nickel nanoparticles. J. Photochem. Photobiol. B Biol. 2018, 178, 143–151. [Google Scholar] [CrossRef] [PubMed]
  252. Sardella, D.; Gatt, R.; Valdramidis, V.P. Assessing the efficacy of zinc oxide nanoparticles against Penicillium expansum by automated turbidimetric analysis. Mycology 2018, 9, 43–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Kolahalam, L.A.; Prasad, K.R.S.; Krishna, P.M.; Supraja, N. Saussurea lappa plant rhizome extract-based zinc oxide nanoparticles: Synthesis, characterization and its antibacterial, antifungal activities and cytotoxic studies against Chinese Hamster Ovary (CHO) cell lines. Heliyon 2021, 7, e07265. [Google Scholar] [CrossRef] [PubMed]
  254. Aziz, N.; Pandey, R.; Barman, I.; Prasad, R. Leveraging the Attributes of Mucor hiemalis-Derived Silver Nanoparticles for a Synergistic Broad-Spectrum Antimicrobial Platform. Front. Microbiol. 2016, 7, 1984. [Google Scholar] [CrossRef]
  255. Dilshad, E.; Bibi, M.; Sheikh, N.A.; Tamrin, K.F.; Mansoor, Q.; Maqbool, Q.; Nawaz, M. Synthesis of Functional Silver Nanoparticles and Microparticles with Modifiers and Evaluation of Their Antimicrobial, Anticancer, and Antioxidant Activity. J. Funct. Biomater. 2020, 11, 76. [Google Scholar] [CrossRef]
  256. Kubin, C.J.; McConville, T.H.; Dietz, D.; Zucker, J.; May, M.; Nelson, B.; Istorico, E.; Bartram, L.; Small-Saunders, J.; Sobieszczyk, M.E.; et al. Characterization of Bacterial and Fungal Infections in Hospitalized Patients with Coronavirus Disease 2019 and Factors Associated with Health Care-Associated Infections. Open Forum Infect. Dis. 2021, 8, ofab201. [Google Scholar] [CrossRef]
  257. Cornely, O.A.; Hoenigl, M.; Lass-Flörl, C.; Chen, S.C.A.; Kontoyiannis, D.P.; Morrissey, C.O.; Thompson III, G.R. Mycoses Study Group Education and Research Consortium (MSG-ERC) and the European Confederation of Medical Mycology (ECMM). Defining breakthrough invasive fungal infection-Position paper of the mycoses study group education and research consortium and the European Confederation of Medical Mycology. Mycoses 2019, 62, 716–729. [Google Scholar] [CrossRef]
  258. Salehi, M.R.; Shahi, F.; Rizvi, F.S.; Ghaderkhani, S.; Zainaldain, H.; Khodavaysi, S.; Jamali-Moghaddam, S.R.; Manshadi, S.A.D.; Rezahosseini, O. Combination antifungal therapy without craniotomy in an immunocompromised patient with rhino-orbito-cerebral mucormycosis: A case report. Caspian J. Intern. Med. 2020, 11, 227–230. [Google Scholar] [CrossRef]
  259. Soffritti, I.; D’Accolti, M.; Fabbri, C.; Passaro, A.; Manfredini, R.; Zuliani, G.; Libanore, M.; Franchi, M.; Contini, C.; Caselli, E. Oral Microbiome Dysbiosis Is Associated with Symptoms Severity and Local Immune/Inflammatory Response in COVID-19 Patients: A Cross-Sectional Study. Front. Microbiol. 2021, 12, 687513. [Google Scholar] [CrossRef]
  260. Zhong, H.; Wang, Y.; Shi, Z.; Zhang, L.; Ren, H.; He, W.; Zhang, Z.; Zhu, A.; Zhao, J.; Xiao, F.; et al. Characterization of respiratory microbial dysbiosis in hospitalized COVID-19 patients. Cell Discov. 2021, 7, 23. [Google Scholar] [CrossRef]
  261. Fan, G.; Cheng, L.; Fu, Z.; Sun, B.; Teng, C.; Jiang, X.; Li, X. Screening of yeasts isolated from Baijiu environments for 2-phenylethanol production and optimization of production conditions. 3 Biotech 2020, 10, 275. [Google Scholar] [CrossRef]
  262. Zhong, W.; Zhang, M.; Li, X.; Zhang, Y.; Wang, Z.; Zheng, J. Enantioselective Resolution of (R, S)-2-Phenoxy-Propionic Acid Methyl Ester by Covalent Immobilized Lipase from Aspergillus oryzae. Appl. Biochem. Biotechnol. 2019, 190, 1049–1059. [Google Scholar] [CrossRef]
  263. Gu, S.; Chen, Y.; Wu, Z.; Chen, Y.; Gao, H.; Lv, L.; Guo, F.; Zhang, X.; Luo, R.; Huang, C.; et al. Alterations of the Gut Microbiota in Patients with Coronavirus Disease 2019 or H1N1 Influenza. Clin. Infect. Dis. 2020, 71, 2669–2678. [Google Scholar] [CrossRef]
  264. Zuo, T.; Liu, Q.; Zhang, F.; Lui, G.C.Y.; Tso, E.Y.; Yeoh, Y.K.; Chen, Z.; Boon, S.S.; Chan, F.K.; Chan, P.K.; et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 2021, 70, 276–284. [Google Scholar] [CrossRef]
  265. Zuo, T.; Zhan, H.; Zhang, F.; Liu, Q.; Tso, E.Y.; Lui, G.C.; Chen, N.; Li, A.; Lu, W.; Chan, F.K.; et al. Alterations in Fecal Fungal Microbiome of Patients with COVID-19 during Time of Hospitalization until Discharge. Gastroenterology 2020, 159, 1302–1310. [Google Scholar] [CrossRef]
  266. Buil, J.B.; Meijer, E.F.; Denning, D.W.; Verweij, P.E.; Meis, J.F. Burden of serious fungal infections in the Netherlands. Mycoses 2020, 63, 625–631. [Google Scholar] [CrossRef]
  267. Mekonnen, Z.K.; Ashraf, D.C.; Jankowski, T.; Grob, S.R.; Vagefi, M.R.; Kersten, R.C.; Simko, J.P.; Winn, B.J. Acute Invasive Rhino-Orbital Mucormycosis in a Patient with COVID-19-Associated Acute Respiratory Distress Syndrome. Ophthalmic Plast. Reconstr. Surg. 2021, 37, e40–e80. [Google Scholar] [CrossRef]
  268. Pakdel, F.; Ahmadikia, K.; Salehi, M.; Tabari, A.; Jafari, R.; Mehrparvar, G.; Rezaie, Y.; Rajaeih, S.; Alijani, N.; Barac, A.; et al. Mucormycosis in patients with COVID-19: A cross-sectional descriptive multicentre study from Iran. Mycoses 2021, 64, 1238–1252. [Google Scholar] [CrossRef]
  269. Placik, D.A.; Taylor, W.L.; Wnuk, N.M. Bronchopleural fistula development in the setting of novel therapies for acute respiratory distress syndrome in SARS-CoV-2 pneumonia. Radiol. Case Rep. 2020, 15, 2378–2381. [Google Scholar] [CrossRef]
  270. Goldman, N.; Fink, D.; Cai, J.; Lee, Y.-N.; Davies, Z. High prevalence of COVID-19-associated diabetic ketoacidosis in UK secondary care. Diabetes Res. Clin. Pr. 2020, 166, 108291. [Google Scholar] [CrossRef]
  271. Singh, A.K.; Singh, R.; Joshi, S.R.; Misra, A. Mucormycosis in COVID-19: A systematic review of cases reported worldwide and in India. Diabetes Metab. Syndr. 2021, 15, 102146. [Google Scholar] [CrossRef]
  272. Singh, K.; Kumar, S.; Shastri, S.; Sudershan, A.; Mansotra, V. Black fungus immunosuppressive epidemic with COVID-19 associated mucormycosis (zygomycosis): A clinical and diagnostic perspective from India. Immunogenetics 2021, 74, 197–206. [Google Scholar] [CrossRef]
  273. Lin, X.; Xu, Y.; Pan, X.; Xu, J.; Ding, Y.; Sun, X.; Song, X.; Ren, Y.; Shan, P.F. Global, regional, and national burden and trend of diabetes in 195 countries and territories: An analysis from 1990 to 2025. Sci. Rep. 2020, 10, 14790. [Google Scholar] [CrossRef]
  274. Drissi, C. Black fungus, the darker side of COVID-19. J. Neuroradiol. 2021, 48, 317–318. [Google Scholar] [CrossRef]
  275. Barron, M.A.; Lay, M.; Madinger, N.E. Surgery and treatment with high-dose liposomal amphotericin B for eradication of craniofacial zygomycosis in a patient with Hodgkin’s disease who had undergone allogeneic hematopoietic stem cell transplantation. J. Clin. Microbiol. 2005, 43, 2012–2014. [Google Scholar] [CrossRef] [Green Version]
  276. Çagatay, A.A.; Öncü, S.S.; Çalangu, S.S.; Yildirmak, T.T.; Özsüt, H.H.; Eraksoy, H.H. Rhinocerebral mucormycosis treated with 32 gram liposomal amphotericin B and incomplete surgery: A case report. BMC Infect. Dis. 2001, 1, 22. [Google Scholar] [CrossRef] [Green Version]
  277. Cornely, O.A.; Alastruey-Izquierdo, A.; Arenz, D.; Chen, S.C.; Dannaoui, E.; Hochhegger, B.; Hoenigl, M.; Jensen, H.E.; Lagrou, K.; Lewis, R.E.; et al. Global guideline for the diagnosis and management of mucormycosis: An initiative of the European Confederation of Medical Mycology in cooperation with the Mycoses Study Group Education and Research Consortium. Lancet Infect. Dis. 2019, 19, e405–e421. [Google Scholar] [CrossRef]
  278. Spellberg, B.; Edwards, J., Jr.; Ibrahim, A. Novel Perspectives on Mucormycosis: Pathophysiology, Presentation, and Management. Clin. Microbiol. Rev. 2005, 18, 556–569. [Google Scholar] [CrossRef] [Green Version]
  279. Boelaert, J.R.; Van Cutsem, J.; de Locht, M.; Schneider, Y.J.; Crichton, R.R. Deferoxamine augments growth and pathogenicity of Rhizopus, while hydroxypyridinone chelators have no effect. Kidney Int. 1994, 45, 667–671. [Google Scholar] [CrossRef] [Green Version]
  280. Ferguson, B.J.; Mitchell, T.G.; Moon, R.; Camporesi, E.M.; Farmer, J. Adjunctive hyperbaric oxygen for treatment of rhinocerebral mucormycosis. Rev. Infect. Dis. 1988, 10, 551–559. [Google Scholar] [CrossRef]
  281. Salmanton-García, J.; Sprute, R.; Stemler, J.; Bartoletti, M.; Dupont, D.; Valerio, M.; Garcia-Vidal, C.; Falces-Romero, I.; Machado, M.; de la Villa, S.; et al. COVID-19-Associated Pulmonary Aspergillosis, March–August 2020. Emerg. Infect. Dis. 2021, 27, 1077–1086. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanism of antimicrobial activity of nanoparticles against fungi Nanoparticles inhibit fungal growth by entering the cells through endocytosis and diffusion processes, and their entry will eventually lead to apoptosis. After entering into the cells, nanoparticle inhibits cell wall/membrane synthesis, disruption of energy transduction, production of ROS, enzyme inhibition, and reduced DNA production, DNA damage, and protein damage.
Figure 1. Mechanism of antimicrobial activity of nanoparticles against fungi Nanoparticles inhibit fungal growth by entering the cells through endocytosis and diffusion processes, and their entry will eventually lead to apoptosis. After entering into the cells, nanoparticle inhibits cell wall/membrane synthesis, disruption of energy transduction, production of ROS, enzyme inhibition, and reduced DNA production, DNA damage, and protein damage.
Ijms 23 12526 g001
Figure 2. Fungi penetrating the blood-brain barrier and circulating in the bloodstream. Fungal propagation involves spore inoculation, escaping phagocytosis by macrophages and neutrophils, hyphae germination, growth by taking advantage of the condition of the host attaching to the endothelium using receptors, and damaging the endothelium and eventually causes impairs ion channels and induces apoptosis. This process results in tissue necrosis, thrombus formation, or hemorrhage.
Figure 2. Fungi penetrating the blood-brain barrier and circulating in the bloodstream. Fungal propagation involves spore inoculation, escaping phagocytosis by macrophages and neutrophils, hyphae germination, growth by taking advantage of the condition of the host attaching to the endothelium using receptors, and damaging the endothelium and eventually causes impairs ion channels and induces apoptosis. This process results in tissue necrosis, thrombus formation, or hemorrhage.
Ijms 23 12526 g002
Figure 3. Diabetic condition promotes the growth of mucormycosis in COVID-19 patients by inducing damage to the pancreas. SARS-CoV-2 enters the human body via spike protein available on the envelope which binds with angiotensin-converting enzyme 2 (ACE 2). COVID-19-affected patients develop a dysfunction of the immune system with a decrease in lymphocyte counts and an exponential rise in inflammation. Treatment of COVID-19 patients with immunosuppressant having uncontrolled diabetes mellitus and ketoacidosis are also at major risk for Mucormycosis as it leads to dysfunctional phagocytes causing impaired intracellular killing by oxidative and non-oxidative mechanisms.
Figure 3. Diabetic condition promotes the growth of mucormycosis in COVID-19 patients by inducing damage to the pancreas. SARS-CoV-2 enters the human body via spike protein available on the envelope which binds with angiotensin-converting enzyme 2 (ACE 2). COVID-19-affected patients develop a dysfunction of the immune system with a decrease in lymphocyte counts and an exponential rise in inflammation. Treatment of COVID-19 patients with immunosuppressant having uncontrolled diabetes mellitus and ketoacidosis are also at major risk for Mucormycosis as it leads to dysfunctional phagocytes causing impaired intracellular killing by oxidative and non-oxidative mechanisms.
Ijms 23 12526 g003
Table 1. Antifungal effects of various types of nanoparticles.
Table 1. Antifungal effects of various types of nanoparticles.
S. No.Type of NanoparticlesSizeTarget FungiReferences
(nm)
1AgNPs17.0A. niger, A. flavus, and F. oxysporum[193]
2AgNPs23.0V. dahliae[194]
3AgNPs10–50A. alternata, S. sclerotiorum,
M. phaseolina, R. solani,
B. cinerea, and C. lunata
[195]
4AgNPs50A. flavus and P. chrysogenum[196]
5AgNPs20B. maydis[197]
6AgNPs29R. nigricans[198]
7AgNPs10–20
Colletotrichum sp.,
F. oxysporum, F. acuminatum,
F. tricinctum, F. graminearum,
F. incarnatum, R. solani,
S. sclerotiorum, and
A. alternate
[199]
8AgNPs75 [200]
9AgNPs50A. alternata and C. lunata.
[201]
H. rostratum, F. solani,
F. oxysporum, and A. alternate
10AgNPs10–20 [202]
A. flavus and A. parasiticus
11AgNPs5–30
F. oxysporum, F. moniliform,
F. solani, F. verticillioides,
F. semitectum, A. flavus,
A. terreus, A. niger, A. ficuum,
P. citrinum, P. islandicum,
P. chrysogenum, R. stolonifer,
Phoma, A. alternata, and
A. chlamydospora
[203]
12AgNPs16–20A. alternate, A. niger,
A. nidulans, C. herbarum,
F. moniliforme, Fusarium spp.,
[204]
13AgNPs25–40F. oxysporum, and T. harzianum[205]
14AgNPs10A. flavus, A. niger, A. tereus,
P. notatum, R. olina, F. solani,
F. oxysporum, T. viride,
V. dahlia, and P. spinosum
[206]
15AgNPs15–400Alternaria sp., F. oxysporum,
F. moniliforme, and
F. tricinctum.
[207]
40–90B. cinerea, P. expansum, A. niger,
Alternaria sp., and Rhizopus sp.
[208]
16CuNPs
20–40
17CuNPs
A. flavus, A. fumigates, and F.
oxysporum.
200–500 [202]
18CuNPs A. flavus and A. parasiticus
10 [136]
19CuNPs F. solani, Neofusicoccum sp.,
3–60and F. oxysporum.
[209]
20CuNPs
30–300
A. niger, A terreus, and[210]
21CuNPs A. fumigatus
22
Alternaria spp., A. niger,[211]
Pythium spp., and
22ZnONPs10–25Fusarium spp.
A. alternata, A solani,
F. expansum, and[212]
23ZnONPs10–25Penicilliun sp.
B. albicans, C. glabrata, C. tropicalis[213]
24TiO2 NPs
70–300
Candida sps[213]
25SeNPs
Candida sps
[214]
Candida albicans
Table 2. Microorganisms involved in the pathogenesis of COVID-19.
Table 2. Microorganisms involved in the pathogenesis of COVID-19.
S. No.Place of OrganName of MicroorganismsReference
1OralStreptococcus, Porphyromonas, Abiotrophia, Enterobacter, Neisseria mucosa, Veillonella parvula,
Lactobacillus fermentum, Enterococcus faecalis, Atopobium parvulum, Acinetobacter baumannii,
Prevotella melaninogenica, jejuni, denticola, and oris; Eikenella corrodens; Capnocytophaga sputigena
and gingivalis; and Aggregatibacter aphrophilus), Aspergillus sp., Nakaseomyces sp., and Malassezia
sp., Candida sp., Saccharomyces sp., Epstein–Barr virus, Staphylococcus phage ROSA, Streptococcus
phage EJ-1, phage PH10, Lactobacillus phage phiadh.
[259]
2LungBurkholderiacepacia complex (BCC), Staphylococcus epidermidis, Mycoplasma spp.
(including M. hominis and M. orale)
[260]
Cutaneotricosporon, Issatchenkia, Wallemia, Cladosporium, Alternaria, Dipodascus, Mortierella,
Aspergillus, Naganishia, Diutina, and Candida
[261]
3LungLung alphaherpesvirus 1, rhinovirus B, human orthopneumovirus[262]
4LungAcinetobacter, Chryseobacterium, Burkholderia, Brevundimonas, Sphingobium, Enterobacteriaceae[261]
5GutStreptococcus, Rothia, Actinomyces, Vellionella, Collinsella aerofaciens, Collinsella tanakaei, Streptococcus infantis, Morganella morganii, Candida albicans, Candida auris, Aspergillus flavus, Coprobacillus sp., Clostridium ramosum, Clostridium hathewayi[263,264,265]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gurunathan, S.; Lee, A.R.; Kim, J.H. Antifungal Effect of Nanoparticles against COVID-19 Linked Black Fungus: A Perspective on Biomedical Applications. Int. J. Mol. Sci. 2022, 23, 12526. https://doi.org/10.3390/ijms232012526

AMA Style

Gurunathan S, Lee AR, Kim JH. Antifungal Effect of Nanoparticles against COVID-19 Linked Black Fungus: A Perspective on Biomedical Applications. International Journal of Molecular Sciences. 2022; 23(20):12526. https://doi.org/10.3390/ijms232012526

Chicago/Turabian Style

Gurunathan, Sangiliyandi, Ah Reum Lee, and Jin Hoi Kim. 2022. "Antifungal Effect of Nanoparticles against COVID-19 Linked Black Fungus: A Perspective on Biomedical Applications" International Journal of Molecular Sciences 23, no. 20: 12526. https://doi.org/10.3390/ijms232012526

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop