Back to Journals » International Journal of Nanomedicine » Volume 19
Green Synthesis of Zinc Oxide Nanoparticles: Preparation, Characterization, and Biomedical Applications - A Review
Authors El-Saadony MT , Fang G, Yan S, Alkafaas SS , El Nasharty MA , Khedr SA, Hussien AM, Ghosh S, Dladla M , Elkafas SS, Ibrahim EH, Salem HM , Mosa WF, Ahmed AE, Mohammed DM, Korma SA, El-Tarabily MK, Saad AM, El-Tarabily KA , AbuQamar SF
Received 16 July 2024
Accepted for publication 17 October 2024
Published 3 December 2024 Volume 2024:19 Pages 12889—12937
DOI https://doi.org/10.2147/IJN.S487188
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 3
Editor who approved publication: Prof. Dr. RDK Misra
Mohamed T El-Saadony,1,* Guihong Fang,2,3,* Si Yan,3 Samar Sami Alkafaas,4 Mahmoud A El Nasharty,5 Sohila A Khedr,6 Aya Misbah Hussien,7 Soumya Ghosh,8 Mthokozisi Dladla,9 Sara Samy Elkafas,10,11 Essam H Ibrahim,12,13 Heba Mohammed Salem,14 Walid FA Mosa,15 Ahmed Ezzat Ahmed,12 Dina Mostafa Mohammed,16 Sameh A Korma,17 Marawan K El-Tarabily,18 Ahmed M Saad,19 Khaled A El-Tarabily,20 Synan F AbuQamar20
1Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt; 2School of Public Health, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China; 3Qionghai People’s Hospital, Qionghai, Hainan, 571400, People’s Republic of China; 4Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt; 5Department of Chemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt; 6Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta, 31733, Egypt; 7Biotechnology Department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21531, Egypt; 8Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman; 9Human Molecular Biology Unit (School of Biomedical Sciences), Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa; 10Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Shebin El Kom, Menofia, 32511, Egypt; 11Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia; 12Biology Department, Faculty of Science, King Khalid University, Abha, 61413, Saudi Arabia; 13Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo, 12611, Egypt; 14Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt; 15Plant Production Department (Horticulture-Pomology), Faculty of Agriculture, Saba Basha, Alexandria University, Alexandria, 21531, Egypt; 16Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt; 17Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt; 18Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary; 19Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt; 20Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
*These authors contributed equally to this work
Correspondence: Si Yan, Qionghai People’s Hospital, Qionghai, Hainan, 571400, People’s Republic of China, Email [email protected] Synan F AbuQamar, Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates, Tel +971 3 713 6733, Fax +971 3 713 4927, Email [email protected]
Abstract: Over the last decade, biomedical nanomaterials have garnered significant attention due to their remarkable biological properties and diverse applications in biomedicine. Metal oxide nanoparticles (NPs) are particularly notable for their wide range of medicinal uses, including antibacterial, anticancer, biosensing, cell imaging, and drug/gene delivery. Among these, zinc oxide (ZnO) NPs stand out for their versatility and effectiveness. Recently, ZnO NPs have become a primary material in various sectors, such as pharmaceutical, cosmetic, antimicrobials, construction, textile, and automotive industries. ZnO NPs can generate reactive oxygen species and induce cellular apoptosis, thus underpinning their potent anticancer and antibacterial properties. To meet the growing demand, numerous synthetic approaches have been developed to produce ZnO NPs. However, traditional manufacturing processes often involve significant economic and environmental costs, prompting a search for more sustainable alternatives. Intriguingly, biological synthesis methods utilizing plants, plant extracts, or microorganisms have emerged as ideal for producing ZnO NPs. These green production techniques offer numerous medicinal, economic, environmental, and health benefits. This review highlights the latest advancements in the green synthesis of ZnO NPs and their biomedical applications, showcasing their potential to revolutionize the field with eco-friendly and cost-effective solutions.
Keywords: antimicrobial, antioxidant, green synthesis, nanomedicine, nanoparticle, zinc oxide
Introduction
Zinc (Zn) is essential to human, animal, and plant metabolism, playing a key role in muscles, bones, skin, and brain function.1 As an essential trace mineral, zinc oxide (ZnO) is commonly used in dietary supplements, cosmetics, and medical products. While most commercially available ZnO is synthetically produced, it also naturally occurs in the earth’s crust as the mineral zincite. ZnO is known for being safe and gentle on human skin, making it ideal for various topical applications.1 ZnO nanoparticles (NPs) have diverse applications across multiple industries, including use as photocatalysts,2 ethanol gas sensors,1,3 ultraviolet (UV) light-emitting devices,4,5 and in the pharmaceutical,6 and cosmetics sectors.7 Metal and metal oxide NPs, including ZnO NPs are integral to advancements in textiles, medicine, catalysis, pharmaceuticals, agriculture, heavy industrial consumer products, and antimicrobial testing.8–11 This versatility highlights the significant role of ZnO and its nanoparticulate forms in driving innovation and improving products that touch our daily lives.9–11
ZnO NPs offer a unique blend of safety, affordability, and versatility, making them a standout among metal oxide NPs.12 Their strong UV absorption and transparency to visible light make them highly effective as sunblock agents.13 In addition, their ability to generate reactive oxygen species (ROS) has spurred research into their antibacterial and anticancer properties.14 ZnO NPs are increasingly recognized for their biomedical applications, particularly in drug delivery systems and bioimaging.15 They can target and release drugs at specific sites, enhancing therapeutic outcomes, and their bioimaging capabilities improve the visualization of biological processes and disease states.13 The United States Food and Drug Administration (FDA) has approved ZnO in bulk as a generally recognized safe substance, and ZnO NPs larger than 100 nm are deemed to be compatible with biological systems,15 highlighting their potential in pharmaceutical delivery applications. Moreover, ZnO NPs are being explored for wound healing due to their antimicrobial properties and ability to promote tissue regeneration. This multifunctionality positions ZnO NPs as a promising tool in various medical and pharmaceutical applications, driving innovation and improving patient care.15
Recent research highlights the advantages of using biological sources as reducing agents in the synthesis of ZnO NPs.16 This eco-friendly approach is favored for its non-hazardous simplicity, low energy consumption, and cost-effectiveness. Plant-derived compounds, such as terpenoids, alkaloids, polyphenols, saponins, flavanones, and tannins, have been shown to effectively reduce Zn precursors. In addition, plant extracts have demonstrated superior efficacy against bacterial and fungal infections.17 This review provides a comprehensive overview of the latest advancements in the green synthesis of ZnO NPs and explores their promising medical applications.
Transition from Conventional to Green Synthesis of ZnO NPs
ZnO NPs offer a balanced combination of low toxicity and high biodegradability compared to other nanomaterials, such as gold nanoparticles (AuNPs), which are non-biodegradable and can accumulate in tissues.18,19 While liposomes also possess non-toxic and biodegradable characteristics, ZnO NPs provide additional therapeutic benefits due to their antibacterial and anti-inflammatory properties.20 Moreover, ZnO NPs are multifunctional and can selectively generate ROS in cancer cells, a significant advantage in targeted cancer therapy.21 Their potential applications in imaging and diagnostics further enhance their appeal.21
NPs can be generated through biological, chemical, and physical methods.22 However, physical methods are not ideal for large-scale production due to low yield, high energy requirements, and increased input costs.23,24 While chemical methods have become the preferred choice for NP synthesis,22,23,25 they are time-consuming, labor-intensive, and pose hazardous effects on humans and the environment.22,23,25 In addition, chemo-synthesized NPs often exhibit instability and toxicity, limiting their biomedical applications.26,27 Therefore, developing efficient, reliable, safe, and environmentally friendly techniques for NP synthesis is crucial.27
Recently, green biological-mediated approaches have gained attention.28,29 Plants and microorganisms can generate NPs that are safe, environmentally sustainable, and cost-effective.30,31 Biological agents, such as algae, bacteria, fungi, and plants, can serve as solvents and stabilizers, creating a green synthetic pathway for NPs and reducing the toxicity of the end product.32 This ability not only diminishes environmental pollution but also transforms heavy metals (HMs) from industrial wastes into safer compounds. The distinctive biochemical mechanisms of these agents offer novel and untapped avenues for converting inorganic metallic ions into metal NPs.30,33,34 Plants contain diverse metabolites and biomolecules, including proteins, vitamins, coenzyme-based intermediates, phenols, flavonoids, and carbohydrates, which can interact with metal ions and reduce their sizes to the nano range.35 For example, flavonoids, characterized by their polyphenolic structures and multiple hydroxyl (-OH) groups attached to aromatic rings, can donate electrons to reduce metal ions such as Zn2+ and Ag+ to their corresponding metallic NPs. Similarly, phenolic compounds, with their hydroxyl groups, act as electron donors, facilitating the reduction of metal ions.17,36
Once the metal ions are reduced, flavonoids and phenolic compounds further contribute to the stabilization and capping of the NPs.37 The hydroxyl and other functional groups in these compounds form strong interactions, such as hydrogen bonding and van der Waals forces, with the metal surface, preventing NP aggregation and enhancing their stability in suspension.37,38 This capping process not only stabilizes NPs but also improves their biocompatibility, making them suitable for various biomedical applications.38
For instance, the flavonoid quercetin has been shown to effectively synthesize silver nanoparticles (AgNPs).39 Quercetin, with its multiple hydroxyl groups, reduces Ag+ to Ag and subsequently caps the AgNPs, enhancing their stability. Another example involves the use of Aloe vera extract in the green synthesis of ZnO NPs.40 A. vera extract, rich in polyphenols, vitamins, enzymes, and amino acids, acts as both a reducing and capping agent. The polyphenolic compounds, such as flavonoids and tannins, reduce Zn2+ ions from zinc nitrate to ZnO NPs.40,41 Simultaneously, the organic molecules, including polysaccharides and proteins in the extract, cap the NPs, preventing aggregation and ensuring stability.40 This dual action of reduction and capping by A. vera extract not only prevents ZnO NPs from clumping together but also enhances their biocompatibility and dispersibility in aqueous solutions, making this method environmentally friendly and suitable for various applications, including antibacterial and anticancer therapies.41
Similar to plants, green algae (Chlorophyceae), blue-green algae (Cyanophyceae), brown algae (Phaeophyceae), and red algae (Rhodophyceae) have secondary metabolites and demonstrate remarkable efficiency in producing NPs of metals and metal oxides.42–44 Algae contain cytotoxic substances, such as laminarians, terpenoids, and fucoidans, which can combat cancer, inhibit proliferation, and suppress tumors.45–48 Due to their lack of external reducing or capping agents, high energy efficiency, affordability, safety, and simplicity, algae are highly recommended for green NP synthesis in the pharmaceutical and biomedical sectors.30,48
The biological synthesis of NPs using microorganisms has gained interest as a sustainable approach for NP production. Bacteria, which can be easily cultured, generate bioactive molecules in liquid form that convert metal ions into metallic NPs.28,49,50 The cellular mass of bacteria can serve as miniature factories for producing metal oxide NPs.50–52 These “nano-factories” hold great potential in modern nanotechnology, facilitating the production of various types of NPs. Typically, green NPs are derived from live cells, bioactive molecules isolated from biological systems, or cell-free supernatants.53 Although the precise production process is not yet fully understood, essential enzymes have been found to contain amino and carboxylic groups that bind to metal ions and subsequently reduce them into metal NPs.54,55 Biological green approaches use plants, plant extracts, or microorganisms to generate NPs as environmentally friendly alternatives to chemical and physical processes. Utilizing microorganisms in NPs production requires complex procedures for maintaining cell cultures and several purification steps.56
Besides being expensive and time-consuming, common methods like chemical precipitation generate dangerous chemical species that stick to surfaces and harm healthcare applications.56,57 Solvent-based techniques, such as solvothermal,58 hydrothermal,59,60 sol-gel,61,62 chemical precipitation,63 are among the most frequently employed strategies for producing ZnO NPs. Some reactions require air and heat to initiate, while others need a nonreactive environment and employ lethal compounds like hydrogen sulfide (H2S) and toxic stabilizers. These compounds used to stabilize NPs are poisonous and have harmful consequences.64 Hazardous substances generated through chemical processes are surface-absorbable and negatively affect medical applications.64
To address these issues, eco-friendly NP production technology has been developed. Green NPs utilize harmless, non-hazardous, and eco-friendly compounds.56 Gelatin is used as a stabilizing agent when the sol-gel method is modified for producing ZnO NPs.65 These ZnO NPs display a hexagonal (Wurtzite) morphology and range from 30–60 nm in diameter. Studies in the impact of varying oxidation temperatures on the structure of ZnO NPs suggest that gelatin holds significant potential as a stabilizing agent in the sol-gel method for generating ZnO particles at the nanoscale.63,65 Due to the high costs and requirements associated with chemical solvents as reducing agents in conventional methods,66 green methods have been used to synthesize ZnO NPs in a flower-shaped form that has garnered increased attention. ZnO NPs have been exploited in various industrial sectors, including pharmaceuticals, cosmetics, antibacterial products, textiles, and automotive industries.56,67 Recent studies indicate that ZnO NPs have superior antibacterial capabilities compared to microparticles.68
Eco-friendly materials like algae, bacteria, fungi, plant leaf extract, natural materials, and biopolymers, offer several advantages in the production of ZnO NPs by leveraging the inherent qualities of microorganisms and plant, especially for pharmaceutical and medical applications.56,67 ZnO NPs serve as fillers in medicinal products, cosmetics, and medication carriers.69 Compared to AgNPs, ZnO NPs are cost-effective and have a desirable white appearance.70 Plant extract are widely used methods to manufacture ZnO NPs. Aloe barbadensis leaf extract was used for fabricating ZnO NPs, yielding spherical and highly stable particles with sizes ranging from 25–40 nm. Adjusting the quantity of leaf broth solution can modulate particle size.71 Algal-based synthesis is another method, where biological catalysts offer greater specificity and control, allowing customization of NP properties.72,73 Overall, green synthesis techniques for producing ZnO NPs offer sustainable methods utilizing enzymatic, plant, microbial, and algae systems, highlighting their potential for eco-friendly nanomaterials and encouraging further research for diverse applications in biomedical sciences, sensor technology, catalysis, and other fields.73
Biosynthesis of ZnO NPs Using Bacteria
Biological methods offer a promising alternative for synthesizing NPs, presenting clear benefits over other approaches due to their safety, simplicity, non-toxic nature, eco-friendliness, biocompatibility, and cost-effectiveness.74 This process utilizes biologically active materials from microorganisms or plant extracts to create ZnO NPs. These materials serve dual roles in NP synthesis, acting as both reducing and capping agents. Typically, the biosynthesis of ZnO NP synthesis involves a metal precursor, such as soluble salts, into prepared biological extracts.74 The reaction leads to a color change, resulting in ZnO NPs powder.74
Microbial fabrication of ZnO NPs can be classified into intracellular and extracellular synthesis. In intracellular synthesis, Zn precursor molecules are taken up by microbial cells, reduced and then extruded as ZnO NPs.72 For instance, Lactobacillus paracasei from dairy products can produce spherical ZnO NPs intracellularly, while Bacillus licheniformis and Bacillus subtilis can synthesize them extracellularly.72–79
The process begins with bacterial isolation and purification on nutrient agar plates using the serial dilution method, followed by incubation at 35–37°C for 24 hours.73 Pure colonies are then cultured for ZnO NPs production. Each bacterial strain is in nutrient broth (at 35–37°C for 24 hours) under shaking conditions. The supernatant is then used for ZnO NP synthesis, where zinc sulfate (ZnSO4) and sodium hydroxide are mixed with the culture filtrate, followed by heating, microwaving, and cooling to facilitate NP formation.73 The resulting white deposition is then washed, centrifuged, and oven-dried at 40°C for 8 hours to obtain powdered ZnO NPs. The NPs are stored for further study, and the most potent bacterial strain is identified.72,75 Table 1 illustrates the various bacterial strains suitable for environmentally friendly ZnO NPs synthesis.
![]() |
Table 1 Green Synthesis of ZnO NPs Using Bacteria |
Biosynthesis of ZnO NPs Using Fungi
Fungal biomass serves as a readily available and renewable resource for producing nanostructured ZnO using an eco-friendly synthesis method.76 This fungal synthesis technique is similar to microbiological procedures, including the isolation and culture of fungi in typical fungal extract preparations.76 During cultivation, the fungal strain is maintained in a sterile environment at a controlled temperature. After a set period, the liquid medium, free of cells but rich in fungal byproducts, is separated by filtration and centrifugation. This solution is then used to produce ZnO NPs.77
Among various fungi, Aspergillus niger is frequently used as the eco-friendly synthesis of ZnO NPs. Successful syntheses have been achieved using A. niger and Aspergillus terreus, which act as biological reducing and capping agents.75 Biomass from fungal strains such as Fusarium oxysporum, Penicillium citrinum, Aspergillus fumigatus, Aspergillus tubulin, and A. niger, can be cultivated in malt glucose yeast peptone (MGYP) broth. These strains are isolated from soil with high HM contamination, unlike standard strains of A. tubulin, A. fumigatus, F. oxysporum, and P. citrinum. The fungi are grown in 250 mL Erlenmeyer flasks containing 50 mL liquid medium with glucose (15.0 g), yeast extract (1.0 g), (NH4)2SO4 (1.0 g), MgSO47H2O (0.1 g), K2HPO4 (2.0 g), and KH2PO4 (7.0 g) per liter.78 Incubation occurred at 28 ± 2°C and 180 rpm for 5 days. The fungal biomass is then washed and transferred to deionized water for further incubation. After this period, the biomass is filtered, and the cell-free filtrate is used for ZnO NP biosynthesis.78 Each treatment involves mixing 10 mL of 3.0 mM ZnSO4 solution with 10 mL of fungal filtrate, adjusting the pH to 6.5, and incubating in an orbital shaker for 72 h in the dark.78 The formation of NPs is indicated by a whitish precipitate at the flask’s base, which is separated by centrifugation at 10,000 rpm for 10 min. Positive and negative controls are maintained by incubating fungal mycelium with deionized water and ZnSO4 solution, respectively.78
This promising, eco-friendly, and cost-effective approach in nanotechnology faces several limitations and challenges. One significant issue is the extended incubation times required for fungal cultures, which generally grow more slowly than bacterial cultures.79 This necessitates longer periods for NP synthesis and involves extensive trial and error to optimize conditions for maximum yield and desired NP properties.79 In addition, fungal synthesis requires specific environmental conditions, including precise temperature and pH ranges, as well as a balanced supply of nutrients. Deviations from these conditions can lead to suboptimal synthesis or even complete failure of the process.79
Yield variations also pose a challenge, as different fungal strains exhibit varying capabilities for synthesizing ZnO NPs.100,101 These results leads to inconsistencies in yield and NP properties. Moreover, factors such as medium composition, aeration, and light exposure significantly affect the yield and quality of ZnO NPs, making reproducibility difficult.102 These biological variabilities complicate the scaling up of the process for industrial applications.100,102
Post-synthesis, the purification and processing of NPs to remove biological contaminants is often complex and involves multiple steps. Controlling the size and shape of ZnO NPs is crucial for their applications; However, achieving uniformity remains challenging with biological synthesis methods.102
Environmental and health concerns must also be addressed, necessitating strict biosafety measures to prevent contamination and mitigate potential health risks associated with handling live fungal cultures.101 Although the method is environmentally friendly, the disposal of fungal biomass and by-products must be managed carefully to avoid environmental contamination.101,102 Addressing these limitations requires ongoing research and development to optimize conditions, improve yields, and ensure the consistency and scalability of fungal synthesis of ZnO NPs.102 Table 2 summarizes studies on the green production of ZnO NPs utilizing fungi.
![]() |
Table 2 Green Synthesis of ZnO NPs Using Fungi |
Biosynthesis of ZnO NPs Using Plants
The biological synthesis of NPs offers a viable alternative to conventional chemical or physical fabrication techniques.24,56,129 Most studies focus on eco-friendly methods for producing metal and oxide NPs, highlighting the efficiency and safety of plant-based synthesis.130 Factors like pH levels and annealing temperature significantly influence the size and morphology of the ZnO NPs. Similarly, gelatin was used as a stabilizing agent in the sol-gel method to produce hexagonal ZnO NPs, which are calcined at various temperatures.131 Jiménez-Rosado et al132 have produced ZnO NPs from pepper extracts high in polyphenols, finding that green synthesis may yield pure smaller NPs than chemical methods.132
Further studies demonstrated the synthesis of ZnO using plant extracts like Solanum rantonnetii and thyme, with varying calcination temperatures affecting the NPs’ characteristics.133 For instance, thyme-synthesized ZnO NPs may exhibit optimal quality of 450°C.134 Leaf extracts of Turkish pine (Pinus Brutia) were also used, showing that the pH levels can significantly alter the morphology and size of ZnO NPs.135 MuthuKathija et al136 have utilized Pisonia alba leaf extract to produce ZnO NPs with notable ultraviolet–visible (UV-Vis) spectroscopy at 375 nm.
Additional research explored various plant extracts for ZnO NP synthesis, including Vitex negundo, Trifolium pratense, Lagenaria siceraria, and green tea leaves, each demonstrating specific applications from antibacterial properties to supercapacitor potential.137 Other studies highlight the effectiveness of ZnO NPs in treating urinary tract infections and other medical conditions.138 Table 3 depicts plant species suited for ecologically friendly ZnO NPs production.
![]() |
Table 3 Green Synthesis of ZnO NPs Using Plants |
Biosynthesis of ZnO NPs Using Algae
The eco-friendly production of ZnO NPs through algae represents a sustainable method with potential applications across diverse sectors.157,158 Algae, abundant and cost-effective biological resources, present a distinct advantage for synthesizing ZnO NPs due to their inherent capacity to generate bioactive substances.158 This procedure utilizes the bioactive elements in algae, such as pigments and proteins, to reduce Zn precursors and stabilize the resulting NPs. This green method eliminates the need for hazardous chemicals and energy-intensive processes, thus reducing environmental impact.158
In addition, the ZnO NPs produced demonstrate improved compatibility with biological systems, showing promise for various applications in medical settings, agriculture, and environmental remediation.159 The creation of ZnO NPs through green synthesis using algae showcases the seamless fusion of biotechnology and nanotechnology, presenting an innovative and environmentally friendly approach to develop novel nanomaterials.158,159 Table 4 illustrates the types of algae that can be utilized in the successful green synthesis of ZnO NPs.
![]() |
Table 4 Green Synthesis of ZnO NPs Using Algae |
Traditional versus Green Methods in Producing ZnO NPs
Both traditional and green methods have their advantages and challenges in the production of ZnO NPs. Traditional methods offer high yield, consistency, and scalability, but they come with a higher environmental cost.172 In contrast, green methods, while environmentally friendly and sustainable, face challenges related to scalability and consistency. Addressing these challenges through standardization, optimization, and technological integration can enhance the industrial applicability of green methods, making them a viable alternative to traditional approaches.172
Traditional methods, such as chemical precipitation, the sol-gel method, and hydrothermal synthesis, are well-established and widely used in industry.173,174 These methods are highly effective, providing controlled size distribution and high purity of ZnO NPs. For instance, chemical precipitation involves reacting zinc salts with alkaline agents to precipitate ZnO NPs, resulting in fast reaction rates and high output.175 Similarly, the sol-gel method produces uniform and pure ZnO NPs through hydrolysis and condensation of Zn precursors.173 Hydrothermal synthesis, which crystallizes ZnO NPs under high pressure and temperature, allows for excellent control over particle size and morphology.176 However, these traditional methods often require significant energy consumption to maintain precise temperature and pH conditions, contributing to a substantial carbon footprint.172,175
The environmental impact of traditional methods is further compounded by the production of hazardous chemical by-products, necessitating careful waste management.172 Despite these drawbacks, traditional methods are highly scalable, with established protocols and infrastructure that ensure consistent and reproducible properties of ZnO NPs across batches.177 However, the costs associated with energy consumption and waste management remain high.
On the other hand, green methods for producing ZnO NPs emphasize sustainability and environmental friendliness. Prominent green methods include microbial synthesis and the use of waste materials.177 Microbial synthesis employs bacteria, fungi, or algae, harnessing their metabolic activity to biosynthesize ZnO NPs with specific shapes and sizes.177,178 In addition, utilizing waste materials such as agricultural or industrial by-products can yield functional ZnO NPs, promoting sustainable resource use. These green methods generally require lower energy consumption, resulting in a reduced carbon footprint.178 They also minimize hazardous chemical waste by utilizing renewable and biodegradable materials, enhancing their sustainability.178
Despite their environmental benefits, green methods face significant challenges in industrial applicability.179 Maintaining consistency and reproducibility at larger scales is difficult due to the inherent variability in biological materials and processes. To overcome this, standardizing biological materials and developing robust protocols are essential.179 Furthermore, green methods often yield lower outputs compared to traditional methods.180 Optimizing biological conditions and scaling up cultivation processes can help address this issue. Process control in biological systems is another challenge, which can be mitigated by integrating advanced monitoring and control systems.179 Although the initial costs for research and development of green methods can be high, investing in pilot projects and forming public-private partnerships can share the costs and risks, making green methods economically viable.179,180
Applications of ZnO NPs in Biomedicine
ZnO NPs, a newer of cost-effective and less hazardous nanomaterial have garnered significant interest in various biomedical fields, including anticancer, antioxidant, anti-inflammatory, antibacterial, and anti-diabetes applications, as well as bioimaging and drug delivery.181 This section highlights current developments in the biological uses of ZnO NPs. Nanotechnology in medical research has facilitated a deeper understanding of molecular biology, potentially enabling the design of novel therapies for diseases that were previously difficult to target due to size limitations.182 The creation of bio-functional NPs is crucial for biomedical purposes, attracting significant interest from numerous research teams in recent years.183
Various materials and chemical manufacturing techniques for biomedical applications of ZnO NPs are currently under investigation.182,183 Zn, a natural element found in all living organisms, is essential for the metabolic processes of humans, animals, and plants.2 All living organisms require exposure to the biosphere’s normal background levels of Zn. ZnO is extensively used in the medical, pharmaceutical, and cosmetic sectors and is recognized for its beneficial use as a dietary supplement.183 Although inhaling ZnO dust and fumes is generally considered harmless, precautions must be taken to avoid it. Consequently, regulations have been established to limit potential exposure.184 The biological activities of various green ZnO nanoparticles are outlined in Table 5.
![]() |
Table 5 Biological Activities of Different Green ZnO NPs |
Anticancer Activity of ZnO NPs
Cancer treatment has traditionally involved surgery, radiation, and chemotherapy.221–223 However, these treatments often come with severe side effects.224 ZnO NPs have shown promise due to their selective cytotoxicity towards malignant cells in vitro. Their surfaces can be modified to enhance this selective cytotoxicity, leading to the elimination of cancerous cells without harming healthy cells.225 Siddiqi et al226 have reported that ZnO NPs are toxic to both Gram-positive and Gram-negative bacteria and can affect primary human T-cells. ZnO NPs also considered safe to living organisms due to the essential nature of Zn(II) ions for adults. These advantages have spurred research into ZnO NPs for cancer treatment, highlighting their potential as biodegradable and biocompatible nanoplatforms.224
ZnO NPs combat tumors by increasing ROS generation and promoting apoptosis.227 Their electrostatic properties are also beneficial for anticancer effects. Neutral hydroxyl groups attached to ZnO NPs change their surface charge behavior. In a high-pH solution, protons move away from the particle surface, giving the surface oxygen atoms a negative charge. At lower pH values, positively charged zinc hydroxide (ZnOH2+) forms on the particle surface. ZnO NPs have a positive surface charge and an isoelectric point of 6.4–6.75 in healthy conditions.228 Cancer cell membranes, however, have a markedly negative potential and contain many anionic phospholipids, such as phosphatidylserine.229
The positive charge ZnO NPs enhances their interactions with cancer cells, increasing cellular absorption, cytotoxicity, and phagocytosis.230 Studies have shown that NPs are harmless to mature human dermal fibroblasts and arterial endothelial cells but are damaging to metastatic tumor cells, and they increase apoptosis in neural stem cells.231 Nanomedicine based on ZnO NPs offers high biocompatibility, cancer targeting ability, ease of surface functionalization, and drug delivery capability, addressing many of the drawbacks of traditional treatments.225 However, challenges remain, such as the need for biocompatible dispersion techniques and a better understanding of the specific cytotoxic mechanisms.225
Autophagy, a process of cellular self-consumption, can be triggered by stimuli like ROS, dysfunctional organelles, protein aggregation, and certain anticancer drugs. This process can lead to apoptosis in cancer cells by promoting self-degradation.232 Therefore, autophagy plays a crucial role in NP-induced cytotoxicity by enhancing cancer cell viability and activating death pathways.232
Hussein and Mohammed151 have synthesized ZnO NPs using grape (Vitis vinifera) extract and demonstrated their significant inhibitory effect against the bacteria Staphylococcus aureus and Klebsiella pneumoniae. These ZnO NPs have also shown notable cytotoxic effects against MCF-7 and AMGM5 human cancer cell lines,151 highlighting their potential in treating both bacterial infections and cancer. Chandrasekaran et al233 have synthesized ZnO NPs chemically and using plant leaves, and evaluated their anticancer, antidiabetic, and antibacterial properties. The plant-derived green-synthesized ZnO NPs exhibit the highest α-amylase inhibition efficiency and significant cytotoxicity against the MCF7 cell line.233 In terms of antibacterial activity, green-synthesized ZnO NPs showed stronger effects against Salmonella typhi and B. subtilis than to chemically synthesized.233 This underscores the enhanced antimicrobial efficacy of green-synthesized ZnO NPs, making them promising for further applications in cancer treatment and beyond.233 The anticancer mechanisms of ZnO NPs are illustrated in Figure 1.
![]() |
Figure 1 The anticancer mechanisms of ZnO NPs. Abbreviations: ZnO NPs, zinc oxide nanoparticles; EPR, endoplasmic reticulum. |
Delivery of Cancer Drugs Using ZnO NPs
Incorporating ZnO NPs into therapeutic formulations significantly enhances the potential for safer and more efficient cancer therapies. By utilizing NP-based drug delivery to target specific sections of cancer cells, it is feasible to reduce the total quantity of drugs used and minimize unwanted side effects.234 ZnO NPs are preferable to other nanomaterials because they are less toxic and more biodegradable. There is considerable interest in using ZnO NPs for cancer treatments.181 Loading drugs such as doxorubicin (DOX), paclitaxel, curcumin, and baicalin onto ZnO NPs can improve their solubility, toxicity, and distribution within cancer cells.235 Previous studies have suggested that both ROS and autophagy influence ZnO NPs’ cytotoxicity, although the specific mechanisms regulating ROS and autophagy remain unidentified.236
Using ZnO NPs, researchers have explored the mechanisms governing autophagy and the correlation between ROS and autophagy in lung epithelial cells.236 Batool and colleagues have synthesized ZnO NPs using A. barbadensis leaf extract for stabilization and capping purposes.237 They determine the drug loading capacity (LC) and loading efficiency (LE) of un-stabilized and polyethylene glycol (PEG)-ZnO NPs with DOX and gemcitabine (GEM). DOX exhibits better LE at 65% (650 mg/g) and LC 32% (320 mg/g) on ZnO NPs than GEM, which showed LE 30.5% (30 mg/g) and LC 16.25% (162 mg/g).237 Similar findings have been observed for PEG-ZnO NPs, with DOX showing 68 and 35% increases in LE (680 mg/g) and LC (350 mg/g), respectively, compared to GEM, whose LE and LC values increased by 35 (350 mg/g) and 19% (190 mg/g), respectively. Using the 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide (MTT) assay, DOX was chosen to encapsulate NPs to assess their in vitro antiproliferative capability against the triple-negative breast cancer (TNBC) cell line (MDA-MB-231).237
Gomathi et al238 have infused DOX into ZnO NPs using the sol-gel process. Various pH conditions (3.0, 6.0, 8.2, and 10.0) have been used to load the drug with a pH of 6.0 determined to be optimal based on UV and SEM analyses. The in vitro cytotoxicity of DOX, ZnO, and ZnO-DOX against HeLa cells is evaluated using the MTT colorimetric cell viability test. ZnO-DOX cells are suppressed by 99.4% at a concentration of 100 g/mL. These findings provide compelling evidence that green biosynthesized ZnO may function well as a potential nano-drug carrier for the targeted drug delivery system.238 Figure 2 depicts anticancer drug delivery using ZnO NPs in cancer treatment.
![]() |
Figure 2 Anticancer drug delivery of ZnO NPs in cancer treatment. Abbreviation: ZnO NPs, zinc oxide nanoparticles. |
ZnO NPs Cytotoxic Effect on Cancer Cells
ZnO NPs induce the death of cancer cells without harming healthy cells.239 However, before ZnO NPs can be used in medicine, several challenges must be addressed, such as developing biocompatible dispersion methods and understanding the mechanisms behind their selective cytotoxicity.181 Research on the cytotoxic effects of ZnO NPs on mammalian cells is limited, and experts do not agree on the significance of existing findings.181
Studies have shown that ZnO NPs reduce the viability of primary human T cells at concentrations lethal to both Gram-negative and Gram-positive bacteria.240 Despite numerous reports indicating that these NPs do not harm cultured human dermal fibroblasts, they are toxic to cancerous and vascular endothelial cells and induce apoptosis in brain stem cells.137 The size of NPs also influences their impacts on cell survival. Van Giau et al138 found that ZnO NPs measuring 8 nm were more toxic to S. aureus than between 50–70 nm. Youssef et al232 observed an inverse relationship between NP size and toxicity in certain cells, specifically noting that smaller NPs generate more ROS.232 Sudhakaran et al241 demonstrated that ZnO NPs are toxic to neural stem cells regardless of particle size, with toxicity varying based on dosage.241
Achieving precise targeting of ZnO NPs to cancer cells without affecting normal cells remains a significant challenge. It is essential to enhance targeting mechanisms to increase specificity and reduce off-target effects.242 Furthermore, large-scale synthesis of ZnO NPs with consistent quality and reproducibility is crucial for clinical applications, as variability in NP size, shape, and surface properties can impact their therapeutic efficacy and safety. The regulatory pathway for the approval of nanomedicine, including ZnO NPs, is complex and requires comprehensive evaluation of their safety, efficacy, and quality.242
Future research directions should prioritize conducting in-depth toxicological studies to understand the long-term effects of ZnO NPs in vivo, focusing on toxicity mechanisms, biodistribution, and clearance from the body.243 It is also important to explore advanced surface modification techniques that enhance the stability, targeting specificity, and therapeutic efficacy of ZnO NPs. Functionalizing the surface with targeting ligands, polymers, and other biomolecules can significantly improve their performance.243
In addition, investigating the use of ZnO NPs in combination with other treatment modalities, such as immunotherapy, can enhance anticancer efficacy; thus, the synergistic effects with existing treatments should be thoroughly evaluated.243 Bridging the gap between laboratory research and clinical application through well-designed pre-clinical and clinical studies is critical. These studies should aim to establish safety, optimal dosing, and therapeutic protocols for ZnO NP-based treatments.243
Developing personalized nanomedicine strategies that utilize ZnO NPs tailored to individual patient profiles could maximize therapeutic efficacy while minimizing adverse effects.243 Finally, research into novel synthesis methods to produce ZnO NPs with controlled size, shape, and surface properties should prioritize green synthesis approaches that utilize eco-friendly materials and processes, thereby enhancing biocompatibility and reducing environmental impact.243
Targeted NPs offer several therapeutic benefits, such as multidrug conjugation, high payload capacity, adjustable discharge kinetics, selective localization, and the ability to evade multidrug resistance mechanisms.244 Various NP functionalization approaches have been developed to enhance the selectivity and specificity of anti-cancer activity. For instance, modifying the surface of ZnO NPs has significantly improved their ability to target specific cancer cells and increased their resilience over time. Research has focused on altering of ZnO NP surfaces using various biological elements, including proteins, hyaluronan, nucleic acids, peptides, and folic acid.232 This biocompatible coating did not affect the anticancer effectiveness of ZnO NPs but also increased their selective targeting of cancer cells while sparing normal cells.232
Anti-Diabetic Activity of ZnO NPs
Research has explored the anti-diabetic effects of ZnO NPs due to their role in facilitating insulin storage, production, and release, given the essential nature of Zn in these processes.245 Studies have shown that ZnO NPs can significantly increase insulin levels, enhance glucose elimination, and improve Zn status, exhibiting higher anti-diabetic activity than ZnSO4.245 For instance, Gadoa et al246 demonstrated that ZnO NPs could restore the cellular structure, improve structural integrity, and normalize biochemical markers such as serum insulin and blood glucose levels, thus proving effective in managing diabetes-induced pancreatic disorders.246 Further trials combined ZnO NPs with diabetic medications like red sandalwood and vildagliptin to enhance efficacy.247 These medications inhibit pancreatic amylase and colonic-glucosidase, enzymes that break down carbohydrates into glucose.248
Diabetes results from a metabolic imbalance due to inadequate insulin production or effectiveness, leading to inefficient carbohydrate processing and persistently high blood sugar levels.249 Inhibiting enzymes like glucosidase and amylase can help regulate blood glucose levels. Current drugs can inhibit these enzymes but often have drawbacks.250 However, Ci-ZnO NPs have demonstrated promising results in suppressing the activities of amylase and glucosidase by interacting with their active and allosteric sites.250 This interaction leads to competitive and non-competitive inhibition, with inhibition percentages ranging between 20–74% for amylase and 36–82% for glucosidase, the latter having a higher inhibition rate.250
For glucosidase, ZnO NPs can bind directly to the active site, preventing substrate access, or to allosteric sites, causing conformational changes that reduce enzyme activity.251 Similarly, for amylase, ZnO NPs can compete with the substrate for the active site or bind allosterically, altering the enzyme’s shape and functionality.251
These interactions result in structural changes, including alterations in the secondary and tertiary structures of the enzymes, which affect their stability and flexibility, ultimately reducing their catalytic efficiency.252 Experimental evidence, including spectroscopic and molecular docking studies, supports these mechanisms, demonstrating that ZnO NPs can induce significant conformational alterations and modify the stability of enzyme molecules.252,253 According to Nazarizadeh and Asri-Rezaie254, over 400 million people worldwide had diabetes in 2014, making it a significant public health concern. Diabetes mellitus arises from metabolic dysfunction where the body either fails to produce enough insulin or utilizes it ineffectively.255 Zn plays a crucial role in insulin storage, synthesis, and secretion and is essential for maintaining insulin’s structural integrity.256 ZnO NPs have been developed as a novel method for Zn administration, and their anti-diabetic benefits have been investigated. For example, red sandalwood extract with ZnO NPs were used as an anti-diabetic medication, finding that the combination was more effective in inhibiting pancreatic glucosidase and amylase than the components alone.233
In another study, Nazarizadeh and Asri-Rezaie254 examined ZnSO4 and ZnO NPs’ antioxidant activity in diabetic rats, finding that small ZnO NPs had a significantly larger antidiabetic impact at higher doses, demonstrated by decreased blood sugar levels, increased insulin levels, and enhanced serum Zn status. Higher doses also aggravated oxidative stress, indicated by increased malondialdehyde (MDA) production and decreased total antioxidant capacity.233
ZnO NPs enhance insulin signaling pathways and intracellular glucose transport primarily by mimicking insulin action and stimulating pancreatic β-cells.257 They activate the insulin receptor, initiating a signaling cascade that includes the phosphorylation of insulin receptor substrates (IRS), activation of phosphatidylinositol-3-kinase (PI3K), and protein kinase B (Akt).258 Akt activation promotes the translocation of glucose transporter 4 (GLUT4) to the cell membrane, facilitating increased glucose uptake.258 In addition, ZnO NPs stimulate β-cells to secrete more insulin and reduce oxidative stress, thereby improving β-cell function and overall glucose homeostasis under diabetic conditions.259
Elevated blood sugar levels can enhance inflammation by modulating the production of C-reactive protein (CRP) and interleukins, which are associated with cardiovascular conditions.249 Rehana et al249 synthesized ZnO NPs using hydroxyethyl cellulose as a stabilizer to alleviate diabetic complications. These NPs reduced levels of asymmetric dimethylarginine (ADMA), fasting blood sugar, MDA, and inflammatory markers like interleukin-1 (IL-1) and CRP in diabetic rats while increasing nitric oxide (NO) levels and antioxidant enzyme PON-1.249
In addition, Eswari et al260 used leaf extract of teak (Tectona grandis) and Indian abutilon (Abutilon indicum) to produce ZnO NPs. The X-ray diffraction (XRD) analysis confirmed the effective creation of wurtzite ZnO NPs with average crystalline sizes of 17 nm for teak and 22 nm for Indian abutilon. Optical assessments revealed absorption bands around the 350 nm UV range, indicating band gap values of 3.0 eV and 3.1 eV.260 The anti-diabetic and anti-inflammatory properties of these ZnO NPs were examined using bovine albumin serum (BSA) denaturation and amylase inhibition techniques, achieving inhibition percentages of 95.42 and 94.82%, respectively.260 Furthermore, MTT tests indicated a reduction in viability among MCF-7 breast cancer cell lines.260 In summary, studies suggest that ZnO NPs hold significant potential for treating diabetes and its complications, due to their ability to improve insulin activity, regulate blood glucose levels, and reduce inflammation.
However, several regulatory challenges must be addressed before ZnO NPs can be integrated into clinical practice. Safety concerns, such as potential cytotoxicity and genotoxicity, require thorough assessment, and long-term exposure studies are necessary to ensure safety.261 Establishing safe and effective dosage ranges is another critical regulatory hurdle.262 In addition, ensuring consistent quality and reproducibility in the production of ZnO NPs is essential for regulatory approval; thus, detailed characterization including size, shape, surface charge, and coating is necessary for regulatory compliance. Ethical considerations, such as informed consent and long-term monitoring of patients, must also be addressed.262
Integrating ZnO NPs into existing diabetes treatment protocols could involve using them as an adjunct therapy alongside current antidiabetic medications, potentially enhancing their effects and reducing required dosages.263,264 Personalized approaches that consider individual patient responses to ZnO NP therapy could be developed for more effective diabetes management. Furthermore, combining ZnO NPs with other therapeutic agents, such as antioxidants, anti-inflammatory drugs, or insulin sensitizers, may provide synergistic benefits.265 While ZnO NPs hold significant promise for diabetes treatment, comprehensive research and rigorous regulatory scrutiny are essential to ensure their safe and effective integration into clinical practice.265
Antimicrobial Activity of ZnO NPs
ZnO NPs exhibit promising antibacterial properties due to their substantial surface area and ability to combat various diseases. Recent studies have highlighted their antimicrobial properties, making them a reliable therapeutic option in medical technology at both micro and nanoscale levels.266–268 Despite their advantages over microparticles, the exact mechanisms underlying their efficacy remain unclear. Notably, ZnO NPs are effective against both Gram-negative and Gram-positive bacteria, as well as spores resistant to high pressure and temperature.226
The concentration and size of ZnO NPs significantly impact their pharmacological behavior. The effectiveness of these NPs correlates with the dimensions and quantity, though the precise therapeutic mechanisms are still not fully understood.269 Some hypotheses suggest that the particle adhesion to microbial surfaces due to constant pressure plays a role, while others propose that the hydrogen peroxide (H2O2) production is a key factor. Increased dosages of ZnO NPs enhance their potency, treatment time, and efficacy,269 partly due to variations in particle size and surface area-to-volume ratio.269
Escherichia coli, Vibrio cholerae, and other Gram-negative bacteria are common models for studying ZnO NPs’ antibacterial effectiveness,270 along with Gram-positive bacteria such as S. aureus.270 Research has also included Proteus vulgaris, Pseudomonas aeruginosa, B. subtilis, and Enterococcus faecalis.70 Studies have shown ZnO NPs exhibit significant antibacterial action against these bacteria, often linked to the production of ROS, which disrupt cell membranes and compromise their integrity.201,271
In specific studies, ZnO with an average size of around 13 nm disrupted bacterial cell membranes through direct contact, while other findings indicated that Zn(II) ions from ZnO NP suspensions did not exhibit antibacterial effects.272 For instance, ZnO NPs inhibited E. coli at approximately 3.4 mM concentrations and S. aureus at concentrations below 1 mM.272 ZnO NPs have also been studied for their potential in treating cholera, a severe intestinal illness caused by V. cholerae.273 Research by Sarwar et al274 revealed that ZnO NPs effectively inhibited the growth of the El Tor (N16961) variant of V. cholerae by inducing ROS overproduction, leading to bacterial membrane damage and increased permeability. In mouse models, ZnO NPs demonstrated the ability to impede cholera toxin’s attachment to the GM1 gangliosides receptor, causing the toxin’s structural collapse.275
The antibacterial effects of ZnO NPs are believed to stem from their ability to generate oxidative stress, disrupt cell membranes, and impede respiratory enzymes through interaction with Zn(I) ions. This leads to ROS and free radical production, causing irreversible damage to bacterial mitochondria, DNA, and membranes.226 NPs can penetrate cell membranes more easily than larger particles, allowing direct interaction with intracellular components, which increases ROS production and subsequent cellular damage.276 The size of NPs also affects their uptake by cells through endocytosis, with smaller ZnO NPs being more readily absorbed and distributed throughout the cytoplasm and organelles, including mitochondria.277 Once inside, these NPs can cause mitochondrial dysfunction, leading to further ROS production and triggering cell death pathways such as apoptosis or necrosis.277,278 The primary mechanism of ZnO NPs toxicity is oxidative stress induced by ROS, with smaller NPs generating higher levels of ROS, overwhelming the cell’s antioxidant defenses.277 This imbalance causes oxidative damage to lipids, proteins, and DNA.277
Elevated ROS levels can also activate inflammatory pathways. Due to their enhanced ROS production, smaller ZnO NPs provoke stronger inflammatory responses, leading to the release of pro-inflammatory cytokines.279 Chronic inflammation can result in tissue damage and contribute to diseases, such as cancer. Furthermore, the ROS generated by smaller ZnO NPs can induce DNA damage, leading to mutations and chromosomal aberrations.280 This genotoxicity may result in cell cycle arrest, apoptosis, or uncontrolled cell proliferation, all of which contribute to carcinogenesis.280
In summary, the toxicity of ZnO NPs is strongly influenced by their size. Smaller NPs, with their higher surface area-to-volume ratio, produce more ROS, leading to increased oxidative stress, cellular damage, and inflammation.64 Understanding these size-dependent effects is crucial for evaluating the safety of ZnO NPs in biomedical and industrial applications. Strategies to mitigate their toxicity may include controlling particle size, surface modifications, or using antioxidants to neutralize ROS.
Studies by Ghasemi and Jalal281 indicated that ZnO NPs enhance the effectiveness of antibiotics like ceftazidime and ciprofloxacin against Acinetobacter baumannii, a pathogen responsible for infections such as pneumonia and meningitis.281 When combined with antibiotics, improved antibiotic absorption and altered bacterial shape, demonstrating the potential for combined treatments.282 ZnO NPs also enhance the antibacterial efficacy of the photosensitizer crystal violet.99,283 Research by Chen et al284 using surface enhanced Raman spectroscopy (SERS) showed that ZnO NPs antibacterial activity varies with dosage and duration, with smaller doses over extended periods mimicking the effects of higher doses.284 The effectiveness of ZnO NPs as antibacterial agents is illustrated in Figure 3.
![]() |
Figure 3 The antibacterial activity of ZnO NPs. Abbreviations: ZnO NPs, zinc oxide nanoparticles; ROS, reactive oxygen species. |
In summary, ZnO NPs possess significant antibacterial properties, and hold promises for various medical and industrial applications. Their dual functionality, including the ability to induce oxidative stress and disrupt cell membranes, makes them potent antibacterial agents suitable for future research and development in combating bacterial infections.
Impact on NPs Modifications on Cellular Uptake and Targeted Interactions with Cancerous Vs Normal Cells
Modifications to NPs significantly influence their cellular uptake and interactions with both cancerous and normal cells.276 These modifications can be optimized to improve targeting, minimize side effects, and enhance therapeutic efficacy.285 Surface modifications play a crucial role in this process. Surface modifications are key, especially when functionalization with targeting ligands such as antibodies, peptides like arginine-glycine-aspartic acid (RGD), aptamers, or small molecules like folic acid. These enable precise targeting of specific antigens overexpressed on cancer cells.285 For instance, HER2 in breast cancer can be targeted with antibodies, while integrins on tumor vasculature can be targeted with RGD peptides.286 Aptamers, which are single-stranded DNA or RNA molecules, bind to specific proteins on cancer cells, and folic acid-conjugated NPs can target folate receptors commonly overexpressed in cancer cells.287
The surface charge of NPs also influences their interaction with cells. Cationic NPs, which are positively charged, interact more readily with the negatively charged cell membranes, enhancing uptake, but may also increase toxicity.288 In contrast, anionic and neutral NPs tend to be less toxic and can be modified with stealth properties, such as PEGylation (attachment of PEG) to evade the immune system and increase circulation time.288,289 The hydrophilicity or hydrophobicity of NP coatings is also crucial; hydrophilic coatings like PEG reduce opsonization and immune recognition290, while hydrophobic coatings enhance interaction with cell membranes but may result in rapid clearance from the bloodstream.291
NPs interaction with cellular receptors is another critical factor. Cancer cells often overexpress receptors, such as transferrin or folate receptors, which can be targeted by corresponding ligands on NPs.292 Mutated receptors in cancer cells present novel targets that normal cells do not have, enabling selective targeting. Normal cells, with tightly regulated receptor expression, provide an opportunity to design NPs that minimize off-target effects by focusing on receptors overexpressed in cancer cells.292
NPs uptake occurs through various cellular pathways. Endocytosis, including clathrin-mediated (CME), caveolae-mediated (CavME), and macropinocytosis, is a primary mechanism for internalization.293 In CME, NPs bind to receptors that cluster into clathrin-coated pits, leading to internalization. CavME involves internalization through caveolin-enriched plasma membrane invaginations, while macropinocytosis allows larger particles or aggregates to be taken up by membrane ruffling and engulfment.294 Small and lipophilic NPs can also passively diffuse through the cell membrane without receptor-mediation.294
Cancerous and normal cells differ significantly in how they interact with and uptake NPs. The enhanced permeability and retention (EPR) effect, characterized by leaky vasculature and poor lymphatic drainage in tumors, allows for passive NP targeting, a feature typically absent in normal tissues.295,296 Tumors also have an acidic microenvironment and higher redox potential compared to normal tissues, which supports the use of pH-sensitive and redox-sensitive NPs for targeted release within the tumor.297 Moreover, hypoxic conditions and the metabolic reprogramming (Warburg effect) in tumors provide further strategies for targeting hypoxia-sensitive and metabolically tuned NPs.298
The Multifaceted Antimicrobial Mechanisms of ZnO NPs
Next-generation nano-antibiotics using ZnO NPs have been developed to combat drug resistance in various treatments.299 These NPs are distinct in their size, crystalline structure, porosity, shape, and content,239 which confer broad antibacterial action against a range of pathogens, including P. aeruginosa, S. aureus, and E. coli.137
In both clinical and non-clinical settings, ZnO NPs can be combined with antibiotics and anti-inflammatory drugs to enhance their effectiveness against harmful microorganisms, while reducing the risk of antibiotic resistance.137,138 Despite the unclear specific mechanisms of their medicinal activity, ZnO is being explored as a pharmacological agent at micro- and nanoscale levels. It is suggested that cell swelling occurs primarily due to ROS generation on particle surfaces, Zn ion release, membrane dysfunction, and NP uptake.300 High temperature processing of ZnO NPs significantly affects their therapeutic efficacy, whereas lower temperature processing has a lesser effect.300 ZnO NPs are also being studied in conjunction with medical ablation techniques and their potential in anti-cancer treatments when exposed to heat.301
ZnO materials exhibit antimicrobial effects through several mechanisms, including the release of Zn(II) ions, adsorption abilities, ROS generation, reactions within microorganisms, induction of lipid peroxidation, interference with DNA replication, and DNA fragmentation,302 Zn(II) ions produced by ZnO NPs/microparticles (MPs) affect metabolic processes and enzyme systems in microorganisms, inducing antibacterial responses.159 Under UV and visible light, ZnO NPs/MPs act as photocatalysts, generating ROS and attracting particles to the bio-membrane through charge interactions.303,304
Furthermore, ZnO NPs can release Zn(II) ions, which interact with the bacterial cell membrane and intracellular components.305,306 These ions bind to negatively charged membrane sites, disrupting membrane potential and inhibiting vital functions like nutrient transport and energy production.305 In addition, ZnO NPs and Zn(II) ions interfere with cellular respiration by inhibiting respiratory enzymes essential for the electron transport chain, leading to reduced ATP production and energy depletion.307,308 Furthermore, ROS generated by ZnO NPs oxidize proteins, disrupting metabolic enzymes and structural proteins crucial for cell integrity.283 ROS can also induce DNA strand breaks, resulting in mutations and impaired replication and transcription.129
ZnO NPs/MPs interact with microorganisms by adsorbing onto their positively charged surfaces and negatively charged cell walls or bio-membranes.309 Their penetration into the membrane causes physical damage, creating pits and perforations that compromise its integrity, leading to leakage and cell death.310 They also mediate lipid peroxidation-induced oxidative stress, which damages DNA. The effectiveness of ZnO NPs/MPs against pathogenic microorganisms varies depending on their porosity, particle size, and shape.309,310
ZnO NPs/MPs exhibit enhanced antibacterial action even against deadly pathogens and show broad antibacterial properties when combined with other antibiotics. This adaptability makes them promising platforms for commercial and clinical applications. Other biomaterials, metal doping, and metal oxide NPs/MPs are also being explored for similar purposes.302
ZnO NPs are not only effective against bacteria but also against fungi, as shown in Figure 4. Abd-Elmaqsoud et al311 found that ZnO NPs derived from Moringa oleifera were detrimental to the plant pathogens, Alternaria saloni and Sclerrotium rolfii. Similarly, Alhazmi and Sharaf312 explored the effectiveness of ZnO NPs against various fungal strains, noting significant impact on both plant and food pathogens, particularly Aspergillus nidulans, Trichoderma harzianum, Rhizopus stoloniferous, and Aspergillus flavus. Consequently, the researchers suggested that ZnO NPs could be utilized in the food and agriculture industries.312 In addition, ZnO NPs exhibit a concentration-dependent effect on the survival of Candida albicans. At a concentration of 0.1 mg/mL, ZnO NPs significantly reduced C. albicans viability. When combined with visible light, the frequency of yeast cell death increased.312
![]() |
Figure 4 The antifungal activity of ZnO NPs. Abbreviation: ZnO NPs, zinc oxide nanoparticles. |
Perveen et al313 produced ZnO NPs using vegetable seed extracts from Brassicaceae plants, including sarson, turnip, white radish, red radish, and cauliflower. These ZnO NPs demonstrated antibacterial activity, with inhibition zones ranging between 10–20 mm, compared to 25 mm for streptomycin.313 This indicates that red radish, white radish, turnip, sarson, and cauliflower can be sources of ZnO NPs with active antibacterial properties.313
The response of bacterial and fungal cell membranes to ZnO NPs differ due to their distinct membrane compositions. In bacterial cells, ZnO NPs primarily disrupt the membrane and increase permeability through electrostatic interactions between the negatively charged membrane components and the positively charged ZnO NPs.314,315 This is further intensified by the generation of ROS, which exacerbates membrane damage, leading to cell lysis and death. In contrast, fungal membranes, rich in sterols like ergosterol, respond to ZnO NPs by increasing membrane permeability and causing cellular contents leakage.316 Although ROS generation also contributes to membrane damage in fungi, the effect is less pronounced due to their stronger antioxidative defenses compared to bacteria.316
Anti-Inflammatory Activity of ZnO NPs
Inflammation is a complex response of body tissues to various potential threats, including irritants, cellular damage, or pathogens.317 The biological functions of Zn ions and the creation of NPs have highlighted the anti-inflammatory capabilities of ZnO NPs. Atopic dermatitis (AD), a chronic inflammatory skin disorder, is characterized by a decreased skin barrier function and a complex interaction between genetic and environmental factors.317
Textiles, which are in close contact with the skin, have been explored for their potential therapeutic effects. Kahru and Mortimer318 investigated the impact of ZnO-enhanced textile fibers on oxidative stress in individuals with AD. Their results indicated that wearing ZnO-induced fabrics overnight for three consecutive days significantly improved pruritus and sleep quality, likely due to the antibacterial and antioxidant properties of ZnO fabrics.318
Youssef et al232 studied the effect of ZnO NPs of varying sizes on allergic skin in a mouse model of AD. They found that the bulk-sized ZnO (bZnO) remained on the skin’s surface layers, while nanoscale ZnO (nZnO) could penetrate the deeper layers of sensitive skin prone to allergic reactions..232 In an animal model of Alzheimer’s disease, nZnO significantly decreased pro-inflammatory cytokines and exerted greater anti-inflammatory effects than bZnO, as evidenced by reductions in Th2 cytokines, IL-13, IFN-, and IL-10.232 Such findings demonstrate that ZnO NPs significantly reduce skin inflammation in AD models.
These interactions primarily occur through the inhibition of key inflammatory signaling pathways. ZnO NPs are taken up by immune cells through endocytosis, where they release Zn ions into the cellular environment.319 These ions disrupt signaling cascades that lead to cytokine production, specifically by reducing the activation of nuclear factor-kappa B (NF-kB), a critical transcription factor involved in pro-inflammatory cytokine expression.213
The NF-κB signaling pathway plays a central role in regulating inflammation and immune responses.320 Normally, NF-κB is kept inactive in the cytoplasm by inhibitor proteins (IκBs).320 Upon activation by pathogens or inflammatory signals, IκB are phosphorylated and degraded, allowing NF-κB to move into the nucleus and trigger the transcription of target genes, including IL-6 and TNF-α.320,321 ZnO NPs inhibit this process by two main mechanisms: First, Zn ions prevent IκB phosphorylation and degradation, thereby blocking NF-κB activation.213 Second, ZnO NPs scavenge ROS, which are involved in NF-κB activation.322 By reducing ROS levels and oxidative stress, ZnO NPs further inhibit NF-κB activation, leading to lower transcription of pro-inflammatory cytokines and reduced inflammation. This modulation of the NF-κB pathway by ZnO NPs highlights their potential as therapeutic agents in controlling inflammation and related diseases.322
The anti-inflammatory capabilities of ZnO NPs extend beyond AD, proving effective for various inflammatory diseases. Imraish et al323 assessed the anti-inflammatory potential of ZnO NPs on RAW 264.7 macrophages stimulated by LPS, noting their superior ability to decrease NO production and suppress the expression of associated proteins such as inducible NO synthase (iNOS), COX-2, IL-1, IL-6, and TNF in a dose-dependent manner.323 This study highlights the broad spectrum of anti-inflammatory effects of ZnO NPs, emphasizing their potential therapeutic applications in various inflammatory conditions.323 Further supporting the anti-inflammatory role of ZnO NPs, Abdelbaky and colleagues324 synthesized ZnO NPs using a solution of Pelargonium odoratissimum leaf extract as a reducing agent.324 In vitro models using the human red blood cells (HRBC) membrane stabilization method (MSM), such as hypotonicity-induced hemolysis, were employed to assess the anti-inflammatory properties of both the ZnO NPs and the water-based extract from P. odoratissimum leaves.324 Compared to conventional indomethacin at a dose of 1000 µg/mL, a maximum membrane stabilization of 95.6% was observed.324
The effectiveness of bulk and nano-ZnO was examined, revealing that only nano-sized ZnO particles can infiltrate the innermost layers of allergic skin, reducing localized skin irritation while promoting the synthesis of IgE antibodies. Ilves et al157 hypothesized that this is due to non-specific processes caused by liberated Zn (II) that limit B-cell IgE synthesis. These findings suggest that ZnO NPs could potentially be used to treat inflammation. The anti-inflammatory impact of ZnO NPs is illustrated in Figure 5.
![]() |
Figure 5 Anti-inflammatory effect of ZnO NPs. Abbreviations: ZnO NPs, zinc oxide nanoparticles; ROS, reactive oxygen species. |
Treatment of Skin Disorders Using ZnO NPs
ZnO is commonly found in medications such as diaper rash powders, barrier creams, antimicrobial ointments, hemimorphite cream, and antidandruff shampoos. Athletes also use ZnO tape to protect soft tissue during exercise.232 Incorporating ZnO NPs into creams, ointments, and lotions provides protection against sunburn and other UV-related skin issues. Due to its excellent UV-A (320–400 nm) and UV-B (280–320 nm) reflectance and perfect photostability, the Bureau has officially approved ZnO for use in sunscreen.232
Studies have shown that ZnO NPs can downregulate type I collagen in skin tissue. Results indicate that ZnO NPs creams at low (1%) and high (6%) concentrations offer preventive benefits against oxidative damage and allergic dermatitis induced by lead oxide, likely due to the anti-inflammatory and antioxidant properties of ZnO NPs.325 At lower concentrations, such as 1%, ZnO NPs can effectively reduce inflammation and oxidative stress, promoting skin health and providing a barrier against environmental pollutants.326 The antioxidant properties neutralize free radicals, reducing oxidative damage to the skin. In addition, their anti-inflammatory effects can mitigate allergic responses and dermatitis by decreasing the production of pro-inflammatory cytokines like IL-6 and TNF-α.327
Conversely, higher concentrations of ZnO NPs, such as 6%, while still offering protection against oxidative stress and inflammation, may pose risks with frequent and prolonged exposure.326 Research shows that in cases of epidermal barrier dysfunction, such as eczema or psoriasis, high concentrations of ZnO NPs increase the risk of adverse effects, including melanoma.322 In mouse models with epidermal barrier failure, topical application of ZnO NPs led to penetration into deeper skin layers, reaching the stratum basale, causing skin lesions resembling melanoma.322 This suggests that high doses of ZnO NPs can disrupt normal cellular functions when the skin barrier is compromised.
Furthermore, both in vitro and in vivo studies have shown that ZnO NPs can exert anti-apoptotic effects on melanocytes by activating the NF-κB pathway through oxidative stress, leading to increased cell survival and proliferation.322 In compromised skin, this could contribute to malignancies like melanoma.322
The distinction between the effects of different ZnO NP concentrations highlights the importance of dosage. While low concentrations primarily offer protective and benefits by reducing oxidative damage and inflammation, high concentrations can pose significant risks, especially in compromised skin conditions.326 The ability of ZnO NPs to penetrate deeper skin layers and activate pathways that prevent apoptosis and promote cell proliferation highlights the need for careful consideration of concentration and exposure duration. Balancing the benefits and risks of ZnO NPs is crucial, particularly in therapeutic and cosmetic applications where long-term skin health is a priority.322,326 The skin-protecting impacts of ZnO NPs are displayed in Figure 6.
![]() |
Figure 6 Skin protective effect of ZnO NPs. Abbreviations: ZnO NPs, zinc oxide nanoparticles; ROS, reactive oxygen species; UV, ultraviolet. |
ZnO NPs and Drug Delivery
Nanotechnology has numerous applications, one of the most important being drug delivery, which has proven effective in treating various diseases, including cancer.328–330 NPs are crucial in medication transportation.221 Many researchers have utilized ZnO NPs for drug delivery across different diseases.181 For instance, Badıllı et al331 employed ZnO quantum dots (QDs) to deliver DOX precisely to HeLa cells. To enhance the stability of these nanomaterials, chitosan was applied to ZnO NPs.181 Their research demonstrated that this drug delivery technology could successfully transport DOX to cancer cells.181 In addition, NPs are significant in gene delivery methods for various cells, particularly malignant cells.181 This gene transfer method offers several advantages, including the effective and secure delivery plasmid-containing genes to target tissues.181
The differences between drug and gene delivery using NPs are significant in terms of mechanisms, requirements, and necessary modifications to ensure efficacy and stability within the body.332 In drug delivery, NPs act as carriers for therapeutic agents like DOX, enabling targeted and controlled release.333 The primary goal is to enhance the drug’s solubility, stability, and bioavailability while minimizing side effects.332,333 For instance, ZnO NPs used for DOX delivery can be modified with chitosan, which not only improves stability but also enhances cellular uptake and controlled release at the target site, ensuring efficient delivery to cancer cells.333
In contrast, gene delivery involves transporting genetic material, such as DNA or RNA, into cells to modify gene expression and treat diseases at the genetic level.334 NPs in gene delivery must protect the genetic material from degradation by nucleases and facilitate its entry into target cells.334,335 Common modifications include adding cationic polymers or lipids to form complexes with negatively charged nucleic acids, protecting them from enzymatic degradation and promoting cellular uptake.336,337 For example, coating NPs with chitosan enhances their ability to bind with DNA or RNA and facilitates endocytosis by target cells.338
Stability within the body is crucial for both drug and gene delivery systems.339 For drug delivery, NPs are often modified with biocompatible and biodegradable materials to prevent premature degradation and ensure sustained release of the therapeutic agent.339–341 In gene delivery, stability is enhanced through protective coatings and stabilizing agents that shield the genetic material from the biological environment.342 In addition, targeting ligands can be attached to the surface of NPs to direct them to specific cell types, improving the efficiency of gene transfer and reducing off-target effects.342
Bio-Imaging and ZnO NPs
This method of gene dispersion has several advantages. Genes generated on the surface of NPs encoded by plasmids can be safely and efficiently transferred to target tissues.343,344 Recent research has explored ZnO nanostructures such as, nanorings, nanorods, nanotubes, and NPs, particularly for their potential in biological imaging.344 This characteristic has several biological and therapeutic uses and applies to varied degrees.344 Luminescent ZnO NPs, also known as ZnO QDs, possess advantageous photophysical properties and are widely recognized for their safety.344 ZnO is used as a food preservative and a component in sunscreen products, making its luminescent properties applicable in various natural and medicinal contexts.344 ZnO is used as fluorescence imaging often used in preclinical research due to its cost-effectiveness and practicability, benefits from these properties.249
Several studies have highlighted the importance of ZnO NPs in cellular imaging. These NPs can emit green fluorescence due to oxygen vacancies and other processes, facilitating the visualization of cancer cells via minimally invasive methods.344 Green, fluorescent ZnO NPs conjugated with transferrin have been used for this purpose. Additionally, the optical properties of ZnO nanomaterials can be modified by incorporating cations like Ni, Cu, or Co, stabilized in aqueous colloidal solutions for various imaging purposes.344 These small ZnO NPs can infiltrate the cell nucleus, and hetero-structural ZnO/Au nanocomposites have been developed and studied for their optical properties and biocompatibility.344
ZnO nanorods can form Au nanoclusters on their tips and surfaces. ZnO nanorods coated with antibodies against the epidermal growth factor receptor have been used to scan cancer cells in vitro.318 QDs are favored for optical imaging due to their attractive optical characteristics.345 ZnO QDs used in in vitro cell imaging showed stable luminescence without cytotoxicity under UV illumination. These QDs have also been evaluated in mice through intradermal and intravenous injections.209
Each imaging method has its own advantages and disadvantages.346 Functionalizing nanomaterials to be detectable by multiple imaging modalities offers synergistic benefits.346 Nanomaterials are more suitable for multimodal imaging than small molecules due to their larger surface areas, providing more sites for functionalization and allowing customization for multimodal detection.347 For instance, sub-6 nm-diameter Gd-doped ZnO QDs have been generated for optical and magnetic resonance imaging (MRI). Fe3O4-ZnO core-shell magnetic QDs have also been investigated for potential cancer imaging and therapy.348
In clinical settings, radionuclide-based imaging techniques like PET and single-photon emission computed tomography (SPECT) are more frequently than optical imaging due to their unlimited tissue penetration, high sensitivity, and quantitative capabilities.346,348 Recent studies have shown that ZnO NPs available in various morphologies, can be used as bioimaging materials.349 Hyperbranched polymers have been employed to produce amphibious ZnO QDs with blue fluorescence, demonstrating their bioimaging applicability.349 ZnO NP surfaces are easily modifiable and durable in aqueous solutions, with water-soluble ZnO enhanced by hyperbranched polyethylenimine compounds performing well in bioimaging.349
Masar et al350 investigated pure n-type ZnO NPs for bioimaging using standard fluorescence microscopy methods. Typically, NPs require UV excitation sources to emit light. However, this study shows that by reducing the energy gap, a 405 nm laser may sufficiently excite NPs for emissions observable during confocal microscope live-cell imaging.350 This research lays the foundation for using these NPs in various bioimaging applications, allowing to study interactions between pristine n-type ZnO NPs and human cells using fluorescence-driven imaging techniques.351 Their developing production process also controls specific defects in pure n-type ZnO NPs for bioimaging.351
ZnO NPs Based Biosensors
Biosensors have broad applications in various fields, including the food industry, healthcare, environmental monitoring, and biological or chemical assessment.352 They are classified based on their detection principles into electrochemical, photometric, piezoelectric, and calorimetric devices.352 Nanomaterials are gaining significant attention due to their unique features, whether used alone or in conjunction with biologically active substances.353 These properties make them a robust foundation for designing high-performance biosensors. The expanded surface area of nanomaterials facilitates the attachment of a wide range of biomolecules, such as antibodies, enzymes, and other proteins.353 Additionally, they enable direct electron transfer from the electrodes to the active regions of the biomolecules.353
ZnO nanomaterials, in particular, have several desirable properties, including high isoelectric point (IEP; 9.5), strong adsorption capacity, excellent biosensing, and high catalytic efficiency.156 These properties make ZnO suitable for electrostatic adsorption of specific proteins like enzymes and antibodies, which have lower IEPs.156 Nanomaterials with higher electron transfer capacities, larger surface areas, and improved biocompatibility or stability are especially beneficial for use in biosensors.354 ZnO-based biosensors are commonly used to detect various small-molecule analytes, such as cholesterol, glucose, H2O2, phenol, and urea. There are numerous biosensors available for detecting specific chemical and physical properties, like pH.355
Dönmez356 created an amperometric glucose biosensor employing ZnO NPs and the root of Zingiber officinale. Glucose oxidase (GOx) was anchored onto a carbon paste electrode (CPE) modified with ZnO through glutaraldehyde cross-linking.298 The resulting biosensor (GOx-ZnO/CPE) demonstrated excellent electrocatalytic glucose measurement capabilities. It featured a low detection limit (14.7 uM), rapid response time (less than 1 second), high sensitivity (15.98 A/mM.cm2), and high biological affinity (Michaelis-Menten constant of 0.99 mM). Additionally, the biosensor showed excellent resistance to interference from uric and ascorbic acids.298
The large surface area and excellent electronic transport properties of ZnO NPs significantly enhance sensor sensitivity by providing more active sites for biomolecule attachment and facilitating efficient electron transfer.348,357 The increased surface area allows for greater adsorption of biomolecules such as enzymes, antibodies, and proteins, improving the chances of interactions with target analytes.358 These interactions are essential for detection, as they enhance the binding affinity and specificity of the sensor.358 In addition, ZnO NPs enable direct electron transfer between the biomolecules and the electrode, leading to faster and more accurate responses.357,358 Together, these combined properties make ZnO NPs highly effective in developing sensitive and reliable biosensors.358
Toxicological Effects of ZnO NPs
The release of Zn-ions from ZnO NPs highlights their potentially hazardous nature, despite being typically considered insoluble in water.359 The pathways through which NPs are taken up by cells influence their surface, shape, size, and properties.359 Enhanced understanding of NP toxicity in both environmental and biological contexts has prompted nano-toxicologists to call for deeper insights into the atomic interactions between NPs and organic structures.359
Aravantinou et al345 investigated the enduring harmful effects of ZnO NPs on microalgae within a reconstructed traditional water treatment setup, incorporating a continuous provision of NPs. Like other metal oxide NPs, ZnO NPs are known for generating ROS and cause apoptosis, in addition to their extraordinary healing capabilities. Due to their properties, ZnO NPs serve as antimicrobial, antibacterial, and anticancer agents.345 They have been shown to produce synergistic benefits when used with various therapy regimens. ZnO NPs are employed for clinical diagnostics and targeted medicine administration, having several applications in the medical field and being environmentally safe.345 Additionally, they are economically viable as they are inexpensive to produce.345
Furthermore, ZnO NPs exhibit differential effects on cancerous versus healthy cells, which is crucial for their safe therapeutic use. In cancerous cells, ZnO NPs induce oxidative stress and apoptosis more effectively due to increased ROS production and an altered redox state.19,360 This targeted effect enhances their potential as anticancer agents. In contrast, healthy cells are generally more resilient to oxidative stress and less susceptible to ZnO NP-induced toxicity.361 However, high concentrations or prolonged exposure can still harm healthy cells, potentially causing inflammation or cellular damage.362 Therefore, precise dosage and targeted delivery are essential to optimize therapeutic benefits while minimizing toxicity risks to healthy tissues.362
Challenges and Future Perspectives
The green synthesis of ZnO NPs faces several challenges that must be overcome for effective biomedical application. A key issue is the variability in biological sources.342 The biochemical compositions of plants, microorganisms, and algae varies significantly, which affects the size, shape, and functionality of the resulting NPs.342 This inconsistency complicates standardization, which is essential for ensuring consistent quality in biomedical applications.342 In addition, scaling up green synthesis from laboratory settings to industrial production remains a significant obstacle.363
Another challenge is the limited understanding of the biochemical mechanisms involved in reducing and stabilizing metal ions through biological agents.359 This knowledge gap makes it difficult to control and optimize the synthesis process, resulting in issues with reproducibility and NP uniformity.359
Regulatory and safety concerns further complicate the application of green synthesis in biomedicine. Although eco-friendly, these methods still require extensive testing for toxicity, biocompatibility, and long-term safety to gain regulatory approval.172 The regulatory landscape for nanomaterials is continually evolving with stringent safety standards necessary for clinical and commercial use.172 Additionally, the cost-effectiveness of green synthesis relies on the availability and accessibility of biological sources.364 Seasonal variations and geographical limitations can impact the supply of raw materials, affecting production and cost.364
Despite these challenges, the future of green synthesis of ZnO NPs is promising. Advancements in characterization techniques, such as high-resolution transmission electron microscopy (HRTEM), X-ray diffraction (XRD), and Fourier-transform infrared spectroscopy (FTIR) are offering deeper insights into the mechanisms of synthesis.365 These tools enable better control over NP production by revealing the interactions between metal ions and biological molecules. Furthermore, integrating green synthesis methods with other eco-friendly technologies like renewable energy sources and biodegradable materials can enhance the sustainability and environmental benefits of ZnO NP production, contributing to a more sustainable nanotechnology industry.342
Functionalizing ZnO NPs with specific biomolecules, ligands, or polymers could enhance targeted delivery in biomedical applications, leading to improved drug delivery systems, better imaging techniques, and more effective therapies.366 Exploring a wider variety of biological sources, such as rare plants, extremophiles, and marine organisms, could also open new avenues for green synthesis of NPs with unique and desirable properties.367
Interdisciplinary collaboration among material scientists, biologists, chemists, and engineers will be critical in overcoming these challenges. Such partnerships can drive innovation and optimize green synthesis methods. Finally, developing comprehensive regulatory frameworks specific to nanomaterials can ensure the safe and sustainable production of ZnO NPs, with clear guidelines to safeguard both environmental and human health.
Conclusion
The green synthesis of ZnO NPs represents a major advancement in biomedical nanotechnology. Over the past decade, their unique properties have been widely explored, particularly in antibacterial treatments, drug and gene delivery, anticancer therapies, cell imaging, and biosensing. While traditional synthetic methods are effective, they pose significant economic and environmental challenges. In contrast, green synthesis method, using plants, plant extracts, and microorganisms, offers a sustainable and eco-friendly alternative that reduces environmental impact, lowers production costs, and minimizes health risks, making it suitable for large-scale production.
Green synthesis harnesses the natural biochemical processes of biological agents, such as plant-derived compounds, to reduce metal ions and stabilize NPs. This method not only adheres to green chemistry principles but also enhances the biocompatibility and therapeutic potential of ZnO NPs. Recent studies emphasize the superior antibacterial and anticancer properties of ZnO NPs produced via green synthesis, highlighting their potential in medical and pharmaceutical applications.
However, several challenges remain before the full benefits of green synthesis can be realized. These include variability in biological sources, difficulties in scaling up production, limited understanding of the underlying biochemical mechanisms, stability concerns, and regulatory barriers. Overcoming these hurdles will require advancements in characterization techniques, integration of green synthesis with other sustainable technologies, and interdisciplinary research efforts. Establishing comprehensive regulatory frameworks is essential for ensuring the safe and effective use of ZnO NPs in biomedical applications.
Despite these obstacles, the future of green synthesis for ZnO NPs is promising. Improved characterization methods, such as HRTEM and FTIR, can provide deeper insights into synthesis mechanisms and better control over production. Functionalizing ZnO NPs with specific biomolecules or polymers can enhance their targeted delivery and therapeutic effectiveness. In addition, exploring a broader range of biological sources may reveal unique properties for NP synthesis. This review highlights the potential of green synthesis to revolutionize nanotechnology, providing eco-friendly, cost-effective solutions that align with global sustainability goals.
Acknowledgments
The authors extend their appreciation to the Deanship of Research and Graduate Studies at King Khalid University for funding this work through Large Research Project under grant number (R.G.P. 2/491/44).
Funding
This work was financially supported by the Finance Science and Technology Project of Hainan Province (ZDYF2019031); UAEU program of Advanced Research (Grant number: 21S169) and Khalifa Center for Biotechnology and Genetic Engineering-UAEU (Grant #: 31R286).
Disclosure
The authors report no conflict of interest in this work.
References
1. Xie J, Cao Y, Jia D, Li Y, Wang Y. Solid-state synthesis of Y-doped ZnO nanoparticles with selective-detection gas-sensing performance. Ceram Int. 2016;42(1):90–96. doi:10.1016/j.ceramint.2015.07.135
2. Sun Y, Zhang W, Li Q, Liu H, Wang X. Preparations and applications of zinc oxide based photocatalytic materials. Adv Sens Energy Mater. 2023;2(3):100069. doi:10.1016/j.asems.2023.100069
3. Adimule V, Revaigh MG, Adarsha HJ. Synthesis and fabrication of Y-doped ZnO nanoparticles and their application as a gas sensor for the detection of ammonia. J Mater Eng Perform. 2020;29:4586–4596. doi:10.1007/s11665-020-04979-4
4. Izu N, Shimada K, Akamatsu T, Itoh T, Shin W, Shiraishi K, Usui T. Polyol synthesis of Al-doped ZnO spherical nanoparticles and their UV–vis–NIR absorption properties. Ceram Int. 2014;40(6):8775–8781. doi:10.1016/j.ceramint.2014.01.099
5. Mishra PN, Mishra PK, Pathak D. The influence of Al doping on the optical characteristics of ZnO nanopowders obtained by the low-cost sol-gel method. Chemistry. 2022;4(4):1136–1146. doi:10.3390/chemistry4040077
6. Lu PJ, Huang SC, Chen YP, Chiueh LC, Shih DY. Analysis of titanium dioxide and zinc oxide nanoparticles in cosmetics. J Food Drug Anal. 2015;23(3):587–594. doi:10.1016/j.jfda.2015.02.009
7. Chen YY, Lee YH, Wang BJ, Chen RJ, Wang YJ. Skin damage induced by zinc oxide nanoparticles combined with UVB is mediated by activating cell pyroptosis via the NLRP3 inflammasome–autophagy–exosomal pathway. Part Fibre Toxicol. 2022;19:2. doi:10.1186/s12989-021-00443-w
8. Akintelu SA,Folorunso AS. A review on green synthesis of zinc oxide nanoparticles using plant extracts and its biomedical applications. BioNanoScience. 2020;10(4):848–863. doi:10.1007/s12668-020-00774-6
9. Fouad OA, Wahsh MMS, Mohamed GG, El Dessouky MMI, Mostafa MR. Modified carbon paste ion selective electrode for determining Cr(iii) ions in aqueous solutions and some real samples using tetragonal zirconia nanoparticles. RSC Adv. 2023;13(16):11201–11214. doi:10.1039/d3ra01563g
10. Fouad OA, Wahsh Mohamed M.S., Mohamed GG, El Desssouky MM, Mostafa MR. Fabrication and characterization of mullite nano-ceramic materials for use in carbon paste ion selective electrode to estimate carcinogenic Cd (II) ion in real and human samples. Microchem J. 2023;190:108623. doi:10.1016/j.microc.2023.108623
11. Fouad OA, Wahsh MM, Mohamed GG, El Desssouky MM. Fabrication and characterization of macroporous alumina-nano tetragonal zirconia-nano spinel ceramic composites. Mater Chem Phys. 2023;301:127617. doi:10.1016/j.matchemphys.2023.127617
12. Al-darwesh MY, Ibrahim SS, Naief MF, Mohammed AM, Chebbi H. Synthesis and characterizations of zinc oxide nanoparticles and its ability to detect O2 and NH3 gases. Results Chem. 2023;6:101064. doi:10.1016/j.rechem.2023.101064
13. Mohammed YH, Holmes A, Haridass IN, Sanchez WY, Studier H, Grice JE, Benson HA, Roberts MS. Support for the safe use of zinc oxide nanoparticle sunscreens: lack of skin penetration or cellular toxicity after repeated application in volunteers. J Invest Dermatol. 2019;139(2):308–315. doi:10.1016/j.jid.2018.08.024
14. Senthilkumar G, Sakthivelu A, Rahman MA, Parameswari P. Enhancement of antibacterial and anticancer properties lanthanum insight into zinc oxide nanoparticles prepared via coprecipitation process. Inorg Chem Commun. 2023;155:111081. doi:10.1016/j.inoche.2023.111081
15. Sahoo RK, Rani S, Kumar V, Gupta U. Zinc oxide nanoparticles for bioimaging and drug delivery. In: Awasthi K, editor. Nanostructured Zinc Oxide. Amsterdam, The Netherlands: Elsevier; 2021:483–509. doi:10.1016/B978-0-12-818900-9.00021-8
16. Khatami M, Alijani HQ, Heli H, Sharifi I. Rectangular shaped zinc oxide nanoparticles: green synthesis by Stevia and its biomedical efficiency. Ceram Int. 2018;44(13):15596–15602. doi:10.1016/j.ceramint.2018.05.224
17. Naseer M, Aslam U, Khalid B, Chen B. Green route to synthesize zinc oxide nanoparticles using leaf extracts of Cassia fistula and Melia azadarach and their antibacterial potential. Sci Rep. 2020;10(1):9055. doi:10.1038/s41598-020-65949-3
18. Niżnik Ł, Noga M, Kobylarz D, Frydrych A, Krośniak A, Kapka-Skrzypczak L, Jurowski K. Gold Nanoparticles (AuNPs)-toxicity, safety and green synthesis: a critical review. Int J Mol Sci. 2024;25(1):4057. doi:10.3390/ijms25074057
19. Anjum S, Hashim M, Malik SA, Khan M, Lorenzo JM, Abbasi BH, Hano C. Recent advances in zinc oxide nanoparticles (ZnO NPs) for cancer diagnosis, target drug delivery, and treatment. Cancers. 2021;13(18):4570. doi:10.3390/cancers13184570
20. Unnikrishnan G, Joy A, Megha M, Kolanthai E, Senthilkumar M. Exploration of inorganic nanoparticles for revolutionary drug delivery applications: a critical review. Discover Nano. 2023;18(1):157. doi:10.1186/s11671-023-03943-0
21. Naser SS, Ghosh B, Simnani FZ, Singh D, Choudhury A, Nandi A, Sinha A, Jha E, Panda PK, Suar M, Verma SK. Emerging trends in the application of green synthesized biocompatible ZnO nanoparticles for translational paradigm in cancer therapy. J Nanotheranostics. 2023;4(3):248–279. doi:10.3390/jnt4030012
22. Koul B, Poonia AK, Yadav D, Jin JO. Microbe-mediated biosynthesis of nanoparticles: applications and future prospects. Biomolecules. 2021;11(6):886. doi:10.3390/biom11060886
23. Varimadugu A, CVS A, Kansoth AN, Mokkapati V, Koodalingam D, Salla S. Microbial synthesis of gold nanoparticles. In: Maddela NR, Díaz JMR, da Silva Montenegro MCB, Prasad R., editors. Microbial Processes for Synthesizing Nanomaterials. Singapore: Springer Nature; 2023:29–59. doi:10.1007/978-981-99-2808-8
24. Shedbalkar U, Singh R, Wadhwani S, Gaidhani S, Chopade BA. Microbial synthesis of gold nanoparticles: current status and future prospects. Adv Colloid Interface Sci. 2014;209:40–48. doi:10.1016/j.cis.2013.12.011
25. Albanese A, Tang PS, Chan WC. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu Rev Biomed Eng. 2012;14:1–16. doi:10.1146/annurev-bioeng-071811-150124
26. You H, Yang S, Ding B, Yang H. Synthesis of colloidal metal and metal alloy nanoparticles for electrochemical energy applications. Chem Soc Rev. 2013;42(7):2880–2904. doi:10.1039/C2CS35319A
27. Shah M, Fawcett D, Sharma S, Tripathy SK, Poinern GEJ. Green synthesis of metallic nanoparticles via biological entities. Materials. 2015;8(11):7278–7308. doi:10.3390/ma8115377
28. Mohd Yusof H, Mohamad R, Zaidan UH, Abdul Rahman NA. Microbial synthesis of zinc oxide nanoparticles and their potential application as an antimicrobial agent and a feed supplement in animal industry: a review. J Anim Sci Biotechnol. 2019;10:57. doi:10.1186/s40104-019-0368-z
29. Salih AM, Al-Qurainy F, Khan S, Tarroum M, Nadeem M, Shaikhaldein HO, Gaafar AZ, Alfarraj NS. Biosynthesis of zinc oxide nanoparticles using Phoenix dactylifera and their effect on biomass and phytochemical compounds in Juniperus procera. Sci Rep. 2021;11(1):19136. doi:10.1038/s41598-021-98607-3
30. El-Sheekh MM, AlKafaas SS, Rady HA, Abdelmoaty BE, Bedair HM, Ahmed AA, El-Saadony MT, AbuQamar SF, El-Tarabily KA. How synthesis of algal nanoparticles affects cancer therapy?–A complete review of the literature. Int J Nanomed. 2023;18:6601–6638. doi:10.2147/IJN.S423171
31. Palaniyandi T, Baskar G, Viswanathan S, Abdul Wahab MR, Govindaraj MK, Sivaji A, Rajendran BK, Kaliamoorthy S. Biosynthesis of iron nanoparticles using brown algae Spatoglossum asperum and its antioxidant and anticancer activities through in vitro and in silico studies. Part Sci Technol. 2023;41(7):916–929. doi:10.1080/02726351.2022.2159900
32. Jha S, Rani R, Singh S. Biogenic zinc oxide nanoparticles and their biomedical applications: a review. J Inorg Organomet Polym Mater. 2023;33(6):1437–1452. doi:10.1007/s10904-023-02550-x
33. El-Saadony MT, Sitohy MZ, Ramadan MF, Saad AM. Green nanotechnology for preserving and enriching yogurt with biologically available iron (II). Innovative Food Sci Emerg Technol. 2021;69:102645. doi:10.1016/j.ifset.2021.102645
34. Saad AM, Sitohy MZ, Sultan-Alolama MI, El-Tarabily KA, El-Saadony MT. Green nanotechnology for controlling bacterial load and heavy metal accumulation in Nile tilapia fish using biological selenium nanoparticles biosynthesized by Bacillus subtilis AS12. Front Microbiol. 2022;13:1015613. doi:10.3389/fmicb.2022.1015613
35. Makarov VV, Love AJ, Sinitsyna OV, Makarova SS, Yaminsky IV, Taliansky ME, Kalinina NO. “Green” nanotechnologies: synthesis of metal nanoparticles using plants. Acta Naturae. 2014;6(20):35–44. doi:10.32607/20758251-2014-6-1-35-44
36. Biswas B, Rogers K, McLaughlin F, Daniels D, Yadav A. Antimicrobial activities of leaf extracts of guava (Psidium guajava L.) on two gram-negative and gram-positive bacteria. Int J Microbiol. 2013;2013:746165. doi:10.1155/2013/746165
37. Sysak S, Czarczynska-Goslinska B, Szyk P, Koczorowski T, Mlynarczyk DT, Szczolko W, Lesyk R, Goslinski T. Metal nanoparticle-flavonoid connections: synthesis, physicochemical and biological properties, as well as potential applications in medicine. Nanomaterials. 2023;13(9):1531. doi:10.3390/nano13091531
38. Nobahar A, Carlier JD, Miguel MG, Costa MC. A review of plant metabolites with metal interaction capacity: a green approach for industrial applications. BioMetals. 2021;34(4):761–793. doi:10.1007/s10534-021-00315-y
39. Reddy NV, Li H, Hou T, Bethu MS, Ren Z, Zhang Z. Phytosynthesis of silver nanoparticles using Perilla frutescens leaf extract: characterization and evaluation of antibacterial, antioxidant, and anticancer activities. Int J Nanomed. 2021;16:15–29. doi:10.2147/IJN.S265003
40. Thongam, DD, Chaturvedi H. Effect of biochemical compounds on ZnO nanomaterial preparation using Aloe vera and lemon extracts. Mater Today. Proc. 2021;44(12):4299–4304. doi:10.1016/j.matpr.2020.10.548
41. P D DA, Plashintania DR, Putri RM, Wibowo I, Ramli Y, Herdianto S, Indarto A. Synthesis of zinc oxide nanoparticles using methanol propolis extract (Pro-ZnO NPs) as antidiabetic and antioxidant. PLoS One. 2023;18(7):e0289125. doi:10.1371/journal.pone.0289125
42. Chaudhary R, Nawaz K, Khan AK, Hano C, Abbasi BH, Anjum S. An overview of the algae-mediated biosynthesis of nanoparticles and their biomedical applications. Biomolecules. 2020;10(11):1498. doi:10.3390/biom10111498
43. El-Sheekh MM, Shabaan MT, Hassan L, Morsi HH. Antiviral activity of algae biosynthesized silver and gold nanoparticles against Herps Simplex (HSV-1) virus in vitro using cell-line culture technique. Int J Environ Health Res. 2022;32(3):616–627. doi:10.1080/09603123.2020.1789946
44. Gheda S, El-Sheekh M, Abou-Zeid A. In vitro anticancer activity of polysaccharide extracted from red alga Jania rubens against breast and colon cancer cell lines. Asian Pac J Trop Med. 2018;11(10):583–589. doi:10.4103/1995-7645.244523
45. Sargazi S, Laraib U, Er S, Rahdar A, Hassanisaadi M, Zafar MN, Díez-Pascual AM, Bilal M. Application of green gold nanoparticles in cancer therapy and diagnosis. Nanomaterials. 2022;12(7):1102. doi:10.3390/nano12071102
46. El-Kassas HY, El-Sheekh MM. Cytotoxic activity of biosynthesized gold nanoparticles with an extract of the red seaweed Corallina officinalis on the MCF-7 human breast cancer cell line. Asian Pac J Cancer Prev. 2014;15(10):4311–4317. doi:10.7314/apjcp.2014.15.10.4311
47. Pitchai P, Subramani P, Selvarajan R, Sankar R, Vilwanathan R, Sibanda T. Green synthesis of gold nanoparticles (AuNPs) using Caulerpa racemosa and evaluation of its antibacterial and cytotoxic activity against human lung cancer cell line. Arab J Basic Appl Sci. 2022;29(1):351–362. doi:10.1080/25765299.2022.2127510
48. de Arruda MC, da Silva MR, Cavalcanti VL, Brandao RM, Marques DD, de Lima LR, Porto AL, Bezerra RP. Antitumor lectins from algae: a systematic review. Algal Res. 2023;70(6):102962. doi:10.1016/j.algal.2022.102962
49. Kitching M, Ramani M, Marsili E. Fungal biosynthesis of gold nanoparticles: mechanism and scale up. Microb Biotechnol. 2015;8(6):904–917. doi:10.1111/1751-7915.12151
50. Mohd Yusof H, Abdul Rahman N, Mohamad R, Zaidan UH, Samsudin AA. Biosynthesis of zinc oxide nanoparticles by cell-biomass and supernatant of Lactobacillus plantarum TA4 and its antibacterial and biocompatibility properties. Sci Rep. 2020;10(1):19996. doi:10.1038/s41598-020-76402-w
51. Moreno-Martin G, Pescuma M, Pérez-Corona T, Mozzi F, Madrid Y. Determination of size and mass-and number-based concentration of biogenic SeNPs synthesized by lactic acid bacteria by using a multimethod approach. Anal Chim Acta. 2017;992:34–41. doi:10.1016/j.aca.2017.09.033
52. Garmasheva I, Kovalenko N, Voychuk S, Ostapchuk A, Livins’ka O, Oleschenko L. Lactobacillus species-mediated synthesis of silver nanoparticles and their antibacterial activity against opportunistic pathogens in vitro. BioImpacts. 2016;6(4):219–223. doi:10.15171/bi.2016.29
53. Ahmed A, Usman M, Ji Z, Rafiq M, Yu B, Shen Y, Cong H. Nature-inspired biogenic synthesis of silver nanoparticles for antibacterial applications. Mater Today Chem. 2023;27:101339. doi:10.1016/j.mtchem.2022.101339
54. Krishnaraj C, Muthukumaran P, Ramachandran R, Balakumaran MD, Kalaichelvan PT. Acalypha indica Linn: biogenic synthesis of silver and gold nanoparticles and their cytotoxic effects against MDA-MB-231, human breast cancer cells. Biotechnol Rep. 2014;4:42–49. doi:10.1016/j.btre.2014.08.002
55. He S, Guo Z, Zhang Y, Zhang S, Wang J, Gu N. Biosynthesis of gold nanoparticles using the bacteria Rhodopseudomonas capsulata. Mater Lett. 2007;61(18):3984–3987. doi:10.1016/j.matlet.2007.01.018
56. Zhou XQ, Hayat Z, Zhang DD, Li MY, Hu S, Wu Q, Cao YF, Yuan Y. Zinc oxide nanoparticles: synthesis, characterization, modification, and applications in food and agriculture. Processes. 2023;11(4):1193. doi:10.3390/pr11041193
57. Ghosh S, Pourebrahimi S, Malloum A, Ajala OJ, AlKafaas SS, Onyeaka H, Nnaji ND, Oroke A, Bornman C, Christian O. Ahmadi S. A review on ciprofloxacin removal from wastewater as a pharmaceutical contaminant: covering adsorption to advanced oxidation processes to computational studies. Mater Today Commun. 2023;107500. doi:10.1016/j.mtcomm.2023.107500
58. Wang Y, Yang C, Liu Y, Fan Y, Dang F, Qiu Y, Zhou H, Wang W, Liu Y. Solvothermal synthesis of ZnO nanoparticles for photocatalytic degradation of methyl Orange and p-nitrophenol. Water. 2021;13(22):3224. doi:10.3390/w13223224
59. Zare M, Namratha K, Thakur MS, Byrappa K. Biocompatibility assessment and photocatalytic activity of bio-hydrothermal synthesis of ZnO nanoparticles by Thymus vulgaris leaf extract. Mater Res Bull. 2019;109:49–59. doi:10.1016/j.materresbull.2018.09.025
60. Ghosh R, Kundu S, Majumder R, Chowdhury MP. Hydrothermal synthesis and characterization of multifunctional ZnO nanomaterials. Mater Today Proc. 2020;26:77–81. doi:10.1016/j.matpr.2019.04.217
61. Haque MJ, Bellah MM, Hassan MR, Rahman S. Synthesis of ZnO nanoparticles by two different methods & comparison of their structural, antibacterial, photocatalytic and optical properties. Nano Res. 2020;1(1):010007. doi:10.1088/2632-959X/ab7a43
62. Arya S, Mahajan P, Mahajan S, Khosla A, Datt R, Gupta V, Young SJ, Oruganti SK. Influence of processing parameters to control morphology and optical properties of sol-gel synthesized Zno nanoparticles. ECS J Solid State Sci Technol. 2021;10(2):023002. doi:10.1088/2632-959X/ab7a43
63. Mahmood NB, Saeed FR, Gbashi KR, Mahmood US. Synthesis and characterization of zinc oxide nanoparticles via oxalate co-precipitation method. Mater Lett X. 2022;13:100126. doi:10.1016/j.mlblux.2022.100126
64. Egbuna C, Parmar VK, Jeevanandam J, Ezzat SM, Patrick-Iwuanyanwu KC, Adetunji CO, Khan J, Onyeike EN, Uche CZ, Akram M, Ibrahim MS. Toxicity of nanoparticles in biomedical application: nanotoxicology. J Toxicol. 2021;2021(1):9954443. doi:10.1155/2021/9954443
65. Babayevska N, Przysiecka Ł, Nowaczyk G, Jarek M, Järvekülg M, Kangur T, Janiszewska E, Jurga S, Iatsunskyi I. Fabrication of gelatin-Zno nanofibers for antibacterial applications. Materials. 2020;14(1):103. doi:10.3390/ma14010103
66. Raha S, Ahmaruzzaman M. ZnO nanostructured materials and their potential applications: progress, challenges and perspectives. Nanoscale Adv. 2022;4(8):1868–1925. doi:10.1039/D1NA00880C
67. Liang SX, Wong LS, Lim YM, Djearamanea S, Lee PF. Effects of zinc oxide nanoparticles on Streptococcus pyogenes. S Afr J Chem Eng. 2020;34(1):63–71. doi:10.1016/j.sajce.2020.05.009
68. Gudkov SV, Burmistrov DE, Serov DA, Rebezov MB, Semenova AA, Lisitsyn AB. A mini-review of antibacterial properties of ZnO nanoparticles. Front Phys. 2021;9:641481. doi:10.3389/fphy.2021.641481
69. Ishwarya R, Vaseeharan B, Kalyani S, Banumathi B, Govindarajan M, Alharbi NS, Kadaikunnan S, Al-Anbr MN, Khaled JM, Benelli G. Facile green synthesis of zinc oxide nanoparticles using Ulva lactuca seaweed extract and evaluation of their photocatalytic, antibiofilm and insecticidal activity. J Photochem Photobiol B. 2018;178:249–258. doi:10.1016/j.jphotobiol.2017.11.006
70. Divya M, Vaseeharan B, Abinaya M, Vijayakumar S, Govindarajan M, Alharbi NS, Kadaikunnan S, Khaled JM, Benelli G. Biopolymer gelatin-coated zinc oxide nanoparticles showed high antibacterial, antibiofilm and anti-angiogenic activity. J Photochem Photobiol B. 2018;178:211–218. doi:10.1016/j.jphotobiol.2017.11.008
71. Youssef FS, Ismail SH, Fouad OA. Mohamed GG.Green synthesis and biomedical applications of zinc oxide nanoparticles. Egypt J Vet Sci. 2023;55(1):287–311. doi:10.21608/EJVS.2023.225862.1576
72. Gahlawat G, Choudhury AR. A review on the biosynthesis of metal and metal salt nanoparticles by microbes. RSC Adv. 2019;9(23):12944–12967. doi:10.1039/C8RA10483B
73. Chandran SP, Chaudhary M, Pasricha R, Ahmad A, Sastry M. Synthesis of gold nanotriangles and silver nanoparticles using Aloe vera plant extract. Biotechnol Prog. 2006;22(2):577–583. doi:10.1021/bp0501423
74. Gomaa EZ. Microbial mediated synthesis of zinc oxide nanoparticles, characterization and multifaceted applications. J Inorg Organomet Polym Mater. 2022;32(11):4114–4132. doi:10.1007/s10904-022-02406-w
75. Chan YY, Pang YL, Lim S, Chong WC. Facile green synthesis of ZnO nanoparticles using natural-based materials: properties, mechanism, surface modification and application. J Environ Chem Eng. 2021;9(4):105417. doi:10.1016/j.jece.2021.105417
76. Ganesan V, Hariram M, Vivekanandhan S, Muthuramkumar S. Periconium sp. (endophytic fungi) extract mediated sol-gel synthesis of ZnO nanoparticles for antimicrobial and antioxidant applications. Mater Sci Semicond Process. 2020;105:104739. doi:10.1016/j.mssp.2019.104739
77. Abdelhakim HK, El-Sayed ER, Rashidi FB. Biosynthesis of zinc oxide nanoparticles with antimicrobial, anticancer, antioxidant and photocatalytic activities by the endophytic Alternaria tenuissima. J Appl Microbiol. 2020;128(6):1634–1646. doi:10.1111/jam.14581
78. Hefny ME, El-Zamek FI, El-Fattah A, Mahgoub SA. Biosynthesis of zinc nanoparticles using culture filtrates of Aspergillus, Fusarium and Penicillium fungal species and their antibacterial properties against gram-positive and gram-negative bacteria. Agric Microbiol Dept Fac Agric Zagazig Univ Egypt. 2019;46(6):2009–2021. doi:10.21608/zjar.2019.51920
79. Morgan RN, Aboshanab KM. Green biologically synthesized metal nanoparticles: biological applications, optimizations and future prospects. Future Sci OA. 2024;10(1):FSO935. doi:10.2144/fsoa-2023-0196
80. Jayaseelan C, Rahuman AA, Kirthi AV, Marimuthu S, Santhoshkumar T, Bagavan A, Gaurav K, Karthik L, Rao KB. Novel microbial route to synthesize ZnO nanoparticles using Aeromonas hydrophila and their activity against pathogenic bacteria and fungi. Spectrochim Acta A. 2012;90:78–84. doi:10.1016/j.saa.2012.01.006
81. Rehman S, Jermy BR, Akhtar S, Borgio JF, Abdul Azeez S, Ravinayagam V, Al Jindan R, Alsalem ZH, Buhameid A, Gani A. Isolation and characterization of a novel thermophile; Bacillus haynesii, applied for the green synthesis of ZnO nanoparticles. Artif Cells Nanomed Biotechnol. 2019;47(1):2072–2082. doi:10.1080/21691401.2019.1620254
82. Tripathi RM, Bhadwal AS, Gupta RK, Singh P, Shrivastav A, Shrivastav BR. ZnO nanoflowers: novel biogenic synthesis and enhanced photocatalytic activity. J Photochem Photobiol B. 2014;141:288–295. doi:10.1016/j.jphotobiol.2014.10.001
83. Saravanan M, Gopinath V, Chaurasia MK, Syed A, Ameen F, Purushothaman N. Green synthesis of anisotropic zinc oxide nanoparticles with antibacterial and cytofriendly properties. Microb Pathog. 2018;115:57–63. doi:10.1016/j.micpath.2017.12.039
84. Alavi M, Kennedy JF. Recent advances of fabricated and modified Ag, Cu, CuO and ZnO nanoparticles by herbal secondary metabolites, cellulose and pectin polymers for antimicrobial applications. Cellulose. 2021;28:3297–3310. doi:10.1007/s10570-021-03746-5
85. Król A, Railean-Plugaru V, Pomastowski P, Złoch M, Buszewski B. Mechanism study of intracellular zinc oxide nanocomposites formation. Colloids Surf A Physicochem Eng. 2018;553:349–358. doi:10.1016/j.colsurfa.2018.05.069
86. Mohd Yusof H, Mohamad R, Zaidan UH, Rahman NA. Sustainable microbial cell nanofactory for zinc oxide nanoparticles production by zinc-tolerant probiotic Lactobacillus plantarum strain TA4. Microb Cell Fact. 2020;19:10. doi:10.1186/s12934-020-1279-6
87. Jemimah VH, Arulpandi I. Evaluation of antimicrobial property of biosynthesized zinc oxide nanoparticles (ZnO NPs) and its application on baby diapers. Drug Invent Today. 2014;6(2):113–119.
88. Barsainya M, Singh DP. Green synthesis of zinc oxide nanoparticles by Pseudomonas aeruginosa and their broad-spectrum antimicrobial effects. J Pure Appl Microbiol. 2018;12(4):2123–2134. doi:10.22207/JPAM.12.4.50
89. Singh BN, Rawat AK, Khan W, Naqvi AH, Singh BR. Biosynthesis of stable antioxidant ZnO nanoparticles by Pseudomonas aeruginosa rhamnolipids. PLoS One. 2014;9(9):e106937. doi:10.1371/journal.pone.0106937
90. Jayabalan J, Mani G, Krishnan N, Pernabas J, Devadoss JM, Jang HT. Green biogenic synthesis of zinc oxide nanoparticles using Pseudomonas putida culture and its in vitro antibacterial and anti-biofilm activity. Biocatal Agric Biotechnol. 2019;21:101327. doi:10.1016/j.bcab.2019.101327
91. Güllüce M, Karadayi M, Demir AB, Isik C, Alaylar B, Ispirli N. Genotoxic potentials of biosynthesized zinc oxide nanoparticles. Pol J Environ Stud. 2020;29(1):111–119. doi:10.15244/pjoes/99239
92. Kundu D, Hazra C, Chatterjee A, Chaudhari A, Mishra S. Extracellular biosynthesis of zinc oxide nanoparticles using Rhodococcus pyridinivorans NT2: multifunctional textile finishing, biosafety evaluation and in vitro drug delivery in colon carcinoma. J Photochem Photobiol B. 2014;140:194–204. doi:10.1016/j.jphotobiol.2014.08.001
93. Shaaban M, El‐Mahdy AM. Biosynthesis of Ag, Se, and ZnO nanoparticles with antimicrobial activities against resistant pathogens using waste isolate Streptomyces enissocaesilis. IET Nanobiotechnol. 2018;12(6):741–747. doi:10.1049/iet-nbt.2017.0213
94. Balraj B, Senthilkumar N, Siva C, Krithikadevi R, Julie A, Potheher IV, Arulmozhi M. Synthesis and characterization of zinc oxide nanoparticles using marine Streptomyces sp. with its investigations on anticancer and antibacterial activity. Res Chem Intermed. 2017;43:2367–2376. doi:10.1007/s11164-016-2766-6
95. Ashour MA, Abd-Elhalim BT. Biosynthesis and biocompatibility evaluation of zinc oxide nanoparticles prepared using Priestia megaterium bacteria. Sci Rep. 2024;14(4147). doi:10.1038/s41598-024-54460-8
96. Suba S, Vijayakumar S, Vidhya E, Punitha VN, Nilavukkarasi M. Microbial mediated synthesis of ZnO nanoparticles derived from Lactobacillus spp: characterizations, antimicrobial and biocompatibility efficiencies. Sens Int. 2021;2:100104. doi:10.1016/j.sintl.2021.100104
97. Hu W, Chen S, Zhou B, Wang H. Facile synthesis of ZnO nanoparticles based on bacterial cellulose. Mater Sci Eng B. 2010;170(1–3):88–92. doi:10.1016/j.mseb.2010.02.034
98. Barani M, Masoudi M, Mashreghi M, Makhdoumi A, Eshghi H. Cell-free extract assisted synthesis of ZnO nanoparticles using aquatic bacterial strains: biological activities and toxicological evaluation. Int J Pharm. 2021;606:120878. doi:10.1016/j.ijpharm.2021.120878
99. Manzoor U, Siddique S, Ahmed R, Noreen Z, Bokhari H, Ahmad I. Antibacterial, structural and optical characterization of mechano-chemically prepared Zno nanoparticles. PLoS One. 2016;11(5):e0154704. doi:10.1371/journal.pone.0154704
100. Kaur T, Bala M, Kumar G, Vyas A. Biosynthesis of zinc oxide nanoparticles via endophyte Trichoderma viride and evaluation of their antimicrobial and antioxidant properties. Arch Microbiol. 2022;204:620. doi:10.1007/s00203-022-03218-9
101. Rani S, Kumar P, Dahiya P, Dang AS, Suneja P. Biogenic synthesis of zinc nanoparticles, their applications, and toxicity prospects. Front Microbiol. 2022;13:824427. doi:10.3389/fmicb.2022.824427
102. Gupta D, Boora A, Thakur A, Gupta TK. Green and sustainable synthesis of nanomaterials: recent advancements and limitations. Environ Res. 2023;231:116316. doi:10.1016/j.envres.2023.116316
103. Murali M, Gowtham HG, Aiyaz M, Sayyed RZ, Achar RR, Silina E, Elbehairi SE, Kollur SP. Zinc oxide nanoparticles prepared through microbial mediated synthesis for therapeutic applications: a possible alternative for plants. Front Microbiol. 2023;14:1227951. doi:10.3389/fmicb.2023.1227951
104. Agarwal H, Kumar SV, Rajeshkumar S. A review on green synthesis of zinc oxide nanoparticles – an eco-friendly approach. Resource-Effic Technol. 2017;3(4):406–413. doi:10.1016/j.reffit.2017.03.002
105. Raliya R, Tarafdar JC. ZnO nanoparticle biosynthesis and its effect on phosphorous-mobilizing enzyme secretion and gum contents in cluster bean (Cyamopsis tetragonoloba L.). Agric Res. 2013;2(1):48–57. doi:10.1007/s40003-012-0049-z
106. Chandrasekaran R, Gnanasekar S, Seetharaman P, Keppanan R, Arockiaswamy W, Sivaperumal S. Formulation of Carica papaya latex-functionalized silver nanoparticles for its improved antibacterial and anticancer applications. J Mol Liq. 2016;219:232–238. doi:10.1016/j.molliq.2016.03.038
107. Baskar G, Chandhuru J, Fahad KS, Praveen AS. Mycological synthesis, characterization and antifungal activity of zinc oxide nanoparticles. Asian J Pharm Technol. 2013;3(4):142–146.
108. Shamsuzzaman Mashrai, A, Khanam H, Aljawfi RN. Biological synthesis of ZnO nanoparticles using C. albicans and studying their catalytic performance in the synthesis of steroidal pyrazolines. Arab J Chem. 2017;10:S1530–S1536. doi:10.1016/j.arabjc.2013.05.004
109. Mashrai A, Khanam H, Aljawfi RN. Biological synthesis of ZnO nanoparticles using C. albicans and studying their catalytic performance in the synthesis of steroidal pyrazolines. Arab J Chem. 2017;10:S1530–S6. doi:10.1016/j.arabjc.2013.05.004
110. Sharma JL, Dhayal V, Sharma RK. White-rot fungus mediated green synthesis of zinc oxide nanoparticles and their impregnation on cellulose to develop environmentally friendly antimicrobial fibers. 3 Biotech. 2021;11(6):269. doi:10.1007/s13205-021-02840-6
111. Nehru L, Kandasamy GD, Sekar V, Alshehri MA, Panneerselvam C, Alasmari A, Kathirvel P. Green synthesis of ZnO-NPs using endophytic fungal extract of Xylaria arbuscula from Blumea axillaris and its biological applications. Artif Cells Nanomed Biotechnol. 2023;51(1):318–333. doi:10.1080/21691401.2023.2232654
112. Jain N, Bhargava A, Tarafdar JC, Singh SK, Panwar J. A biomimetic approach towards synthesis of zinc oxide nanoparticles. Appl Microbiol Biotechnol. 2013;97(2):859–869. doi:10.1007/s00253-012-3934-2
113. Es-haghi A, Soltani M, Karimi E, Namvar F, Homayouni-Tabrizi M. Evaluation of antioxidant and anticancer properties of zinc oxide nanoparticles synthesized using Aspergillus niger extract. Mater Res Express. 2019;6(12):125415. doi:10.1088/2053-1591/ab5f72
114. Kalpana V, Kataru BAS, Sravani N, Vigneshwari T, Panneerselvam A, Rajeswari VD. Biosynthesis of zinc oxide nanoparticles using culture filtrates of Aspergillus niger: antimicrobial textiles and dye degradation studies. OpenNano. 2018;3:48–55. doi:10.1016/j.onano.2018.06.001
115. Ibrahem EJ, Thalij KM, Saleh MK, Badawy AS. Biosynthesis of zinc oxide nanoparticles and assay of antibacterial activity. Am J Biochem Biotechnol. 2017;13(2):63–69. doi:10.3844/ajbbsp.2017.63.69
116. Gao Y, Arokia Vijaya Anand M, Ramachandran V, Karthikkumar V, Shalini V, Vijayalakshmi S, Ernest D. Biofabrication of zinc oxide nanoparticles from Aspergillus niger, their antioxidant, antimicrobial and anticancer activity. J Clust Sci. 2019;30:937–946. doi:10.1007/s10876-019-01551-6
117. Li JF, Rupa EJ, Hurh J, Huo Y, Chen L, Han Y, Chan Ahn J, Park JK, Lee HA, Mathiyalagan R, Yang DC. Cordyceps militaris fungus mediated zinc oxide nanoparticles for the photocatalytic degradation of methylene blue dye. Optik. 2019;183:691–697. doi:10.1016/j.ijleo.2019.02.081
118. Mohamed AA, Fouda A, Abdel-Rahman MA, Hassan SE, El-Gamal MS, Salem SS, Shaheen TI. Fungal strain impacts the shape, bioactivity and multifunctional properties of green synthesized zinc oxide nanoparticles. Biocatal Agric Biotechnol. 2019;19:101103. doi:10.1016/j.bcab.2019.101103
119. Fouda A, Salem SS, Wassel AR, Hamza MF, Shaheen TI. Optimization of green biosynthesized visible light active CuO/ZnO nano-photocatalysts for the degradation of organic methylene blue dye. Heliyon. 2020;6(9):e04896. doi:10.1016/j.heliyon.2020.e04896
120. Singh A, Dutta PK. Green synthesis, characterization and biological evaluation of chitin glucan based zinc oxide nanoparticles and its curcumin conjugation. Int J Biol Macromol. 2020;156:514–521. doi:10.1016/j.ijbiomac.2020.04.081
121. Shobha B, Lakshmeesha TR, Ansari MA, Almatroudi A, Alzohairy MA, Basavaraju S, Alurappa R, Niranjana SR, Chowdappa S. Mycosynthesis of ZnO nanoparticles using Trichoderma spp. isolated from rhizosphere soils and its synergistic antibacterial effect against Xanthomonas oryzae pv. Oryzae J Fungi. 2020;6(3):181. doi:10.3390/jof6030181
122. Sumanth B, Lakshmeesha TR, Ansari MA, Alzohairy MA, Udayashankar AC, Shobha B, Niranjana SR, Srinivas C, Almatroudi A. Mycogenic synthesis of extracellular zinc oxide nanoparticles from Xylaria acuta and its nanoantibiotic potential. Int J Nanomed. 2020;15:8519–8536. doi:10.2147/IJN.S271743
123. Ameen F, Dawoud T, AlNadhari S. Ecofriendly and low-cost synthesis of ZnO nanoparticles from Acremonium potronii for the photocatalytic degradation of azo dyes. Environ Res. 2021;202:111700. doi:10.1016/j.envres.2021.111700
124. Mohamed AA, Abu-Elghait M, Ahmed NE, Salem SS. Eco-friendly mycogenic synthesis of ZnO and CuO nanoparticles for in vitro antibacterial, antibiofilm, and antifungal applications. Biol Trace Elem Res. 2021;199(7):2788–2799. doi:10.1007/s12011-020-02369-4
125. Mousa SA, El-Sayed ER, Mohamed SS, Abo El-Seoud MA, Elmehlawy AA, Abdou DAM. Novel mycosynthesis of Co3 O4, CuO, Fe3O4, NiO, and ZnO nanoparticles by the endophytic Aspergillus terreus and evaluation of their antioxidant and antimicrobial activities. Appl Microbiol Biotechnol. 2021;105(2):741–753. doi:10.1007/s00253-020-11046-4
126. Mekky AE, Farrag AA, Hmed AA, Sofy AR. Preparation of zinc oxide nanoparticles using Aspergillus niger as antimicrobial and anticancer agents. J Pure Appl Microbiol. 2021;15(3):1547–1566. doi:10.22207/JPAM.15.3.49
127. Rasha E, Alkhulaifi MM, AlOthman M, Khalid I, Doaa E, Alaa K, Awad MA. Abdalla M. Effects of zinc oxide nanoparticles synthesized using Aspergillus niger on carbapenem-resistant Klebsiella pneumonia in vitro and in vivo. Front Cell Infect Microbiol. 2021;11:748739. doi:10.3389/fcimb.2021.748739
128. Es-Haghi A, Taghavizadeh Yazdi ME, Sharifalhoseini M, Baghani M, Yousefi E, Rahdar A, Baino F. Application of response surface methodology for optimizing the therapeutic activity of ZnO nanoparticles biosynthesized from Aspergillus niger. Biomimetics. 2021;6(2):34. doi:10.3390/biomimetics6020034
129. Maynard S, Schurman SH, Harboe C, de Souza-Pinto NC, Bohr VA. Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis. 2009;30(1):2–10. doi:10.1093/carcin/bgn250
130. Taccola L, Raffa V, Riggio C, Vittorio O, Iorio MC, Vanacore R, Pietrabissa A, Cuschieri A. Zinc oxide nanoparticles as selective killers of proliferating cells. Int J Nanomed. 2011;6:1129–1140. doi:10.2147/IJN.S16581
131. Kc B, Paudel SN, Rayamajhi S, Karna D, Adhikari S, Shrestha BG, Bisht G. Enhanced preferential cytotoxicity through surface modification: synthesis, characterization and comparative in vitro evaluation of TritonX-100 modified and unmodified zinc oxide nanoparticles in human breast cancer cell (MDA-MB-231). Chem Cent J. 2016;10:16. doi:10.1186/s13065-016-0162-3
132. Jiménez-Rosado M, Gomez-Zavaglia A, Guerrero A, Romero A. Green synthesis of ZnO nanoparticles using polyphenol extracts from pepper waste (Capsicum annuum). J Clean Prod. 2022;350:131541. doi:10.1016/j.jclepro.2022.131541
133. Salem NM, Awwad AM. Green synthesis and characterization of ZnO nanoparticles using Solanum rantonnetii leaves aqueous extract and antifungal activity evaluation. Chem Int. 2022;8(1):12–17. doi:10.5281/zenodo.6848928
134. Karam ST. Abdulrahman AF. Green synthesis and characterization of ZnO nanoparticles by using thyme plant leaf extract. Photonics. 2022;9(8):594. doi:10.3390/photonics9080594
135. Ismail SMM, Ahmed SM, Abdulrahman AF, AlMessiere MA. Characterization of green synthesized of ZnO nanoparticles by using Pinus brutia leaves extracts. J Mol Struct. 2023;1280:135063. doi:10.1016/j.molstruc.2023.135063
136. MuthuKathija M, Badhusha MS, Rama V. Green synthesis of zinc oxide nanoparticles using Pisonia alba leaf extract and its antibacterial activity. Appl Surf Sci Adv. 2023;15:100400. doi:10.1016/j.apsadv.2023.100400
137. Sánchez-López E, Gomes D, Esteruelas G, Bonilla L, Lopez-Machado AL, Galindo R, Cano A, Espina M, Ettcheto M, Camins A, Silva AM, Durazzo A, Santini A, Garcia ML, Souto EB. Metal-based nanoparticles as antimicrobial agents: an overview. Nanomaterials. 2020;10(2):292. doi:10.3390/nano10020292
138. Van Giau V, An SS, Hulme J. Recent advances in the treatment of pathogenic infections using antibiotics and nano-drug delivery vehicles. Drug Des Dev Ther. 2019;13:327–343. doi:10.2147/DDDT.S190577
139. Jayachandran A, Nair AS. Green synthesis and characterization of zinc oxide nanoparticles using Cayratia pedata leaf extract. Biochem Biophys Rep. 2021;26:100995. doi:10.1016/j.bbrep.2021.100995
140. Arumugam J, Thambidurai S, Suresh S, Selvapandiyan M, Kandasamy M, Pugazhenthiran N, Kumar SK, Muneeswaran T, Quero F. Green synthesis of zinc oxide nanoparticles using Ficus carica leaf extract and their bactericidal and photocatalytic performance evaluation. Chem Phys Lett. 2021;783:139040. doi:10.1016/j.cplett.2021.139040
141. Ashwini J, Aswathy TR, Rahul AB, Thara GM, Nair AS. Synthesis and characterization of zinc oxide nanoparticles using Acacia caesia bark extract and its photocatalytic and antimicrobial activities. Catalysts. 2021;11(12):1507. doi:10.3390/catal11121507
142. Kpega TC, Habila JD, Okon IE, Ekwumemgbo PA. Green synthesis and characterization of zinc oxide nanoparticles using Corchorus olitorius leaf extract. Aceh Int J Sci Tech. 2023;12(3):358–367. doi:10.13170/aijst.12.3.34013
143. Abel S, Tesfaye JL, Shanmugam R, Dwarampudi LP, Lamessa G, Nagaprasad N, Benti M, Krishnaraj R. Green synthesis and characterizations of zinc oxide (ZnO) nanoparticles using aqueous leaf extracts of coffee (Coffea arabica) and its application in environmental toxicity reduction. J Nanomater. 2021;2021(1):1–6. doi:10.1155/2021/3413350
144. Selvanathan V, Aminuzzaman M, Tan LX, Win YF, Cheah ES, Heng MH, Tey LH, Arullappan S, Algethami N, Alharthi SS, Sultana S. Synthesis, characterization, and preliminary in vitro antibacterial evaluation of ZnO nanoparticles derived from soursop (Annona muricata L.) leaf extract as a green reducing agent. J Mater Res Technol. 2022;20:2931–2941. doi:10.1016/j.jmrt.2022.08.028
145. Zhu W, Hu C, Ren Y, Lu Y, Song Y, Ji Y, Han C, He J. Green synthesis of zinc oxide nanoparticles using Cinnamomum camphora (L.) Presl leaf extracts and its antifungal activity. J Environ Chem Eng. 2021;9(6):106659. doi:10.1016/j.jece.2021.106659
146. Sana SS, Vadde R, Kumar R, Arla SK, Somala AR, Rao KK, Zhijun Z, Boya VK, Mondal K, Mamidi N. Eco-friendly and facile production of antibacterial zinc oxide nanoparticles from Grewia flavescens (G. flavescens) leaf extract for biomedical applications. J Drug Deliv Sci Technol. 2023;80:104186. doi:10.1016/j.jddst.2023.104186
147. Gaur J, Vikrant K, Kim KH, Kumar S, Pal M, Badru R, Masand S, Momoh J. Photocatalytic degradation of Congo red dye using zinc oxide nanoparticles prepared using Carica papaya leaf extract. Mater Today Sustain. 2023;22:100339. doi:10.1016/j.mtsust.2023.100339
148. Álvarez-Chimal R, García-Pérez VI, Álvarez-Pérez MA Tavera-Hernández R, Reyes-Carmona L, Martínez-Hernández M, Arenas-Alatorre JÁ. Influence of the particle size on the antibacterial activity of green synthesized zinc oxide nanoparticles using Dysphania ambrosioides extract, supported by molecular docking analysis. Arabian J Chem. 2022;15(6):103804. doi:10.1016/j.arabjc.2022.103804
149. Fouda A, Saied E, Eid AM, Kouadri F, Alemam AM, Hamza MF, Alharbi M, Elkelish A, Hassan SE. Green synthesis of zinc oxide nanoparticles using an aqueous extract of Punica granatum for antimicrobial and catalytic activity. J Funct Biomater. 2023;14(4):205. doi:10.3390/jfb14040205
150. Safavinia L, Akhgar MR, Tahamipour B, Ahmadi SA. Green synthesis of highly dispersed zinc oxide nanoparticles supported on silica gel matrix by Daphne oleoides extract and their antibacterial activity. Iran J Biotechnol. 2021;19(1):e2598. doi:10.30498/IJB.2021.2598
151. Hussein BY, Mohammed AM. Green synthesis of ZnO nanoparticles in grape extract: their application as anti-cancer and anti-bacterial. Mater Today Proc. 2021;42:A18–A26. doi:10.1016/j.matpr.2021.03.729
152. Nilavukkarasi M, Vijayakumar S, Prathipkumar S. Capparis zeylanica mediated bio-synthesized ZnO nanoparticles as antimicrobial, photocatalytic and anti-cancer applications. Mater Sci Energy Technol. 2020;3:335–343. doi:10.1016/j.mset.2019.12.004
153. Sharmila G, Thirumarimurugan M, Muthukumaran C. Green synthesis of ZnO nanoparticles using Tecoma castanifolia leaf extract: characterization and evaluation of its antioxidant, bactericidal and anticancer activities. Microchem J. 2019;145:578–587. doi:10.1016/j.microc.2018.11.022
154. Jevapatarakul D, T-Thienprasert J, Payungporn S, Chavalit T, Khamwut A, T-Thienprasert NP. Utilization of Cratoxylum formosum crude extract for synthesis of ZnO nanosheets: characterization, biological activities and effects on gene expression of nonmelanoma skin cancer cell. Biomed Pharmacother. 2020;130:110552. doi:10.1016/j.biopha.2020.110552
155. Duan X, Liao Y, Liu T, Yang H, Liu Y, Chen Y, Ullah R, Wu T. Zinc oxide nanoparticles synthesized from Cardiospermum halicacabum and its anticancer activity in human melanoma cells (A375) through the modulation of apoptosis pathway. J Photochem Photobiol B. 2020;202:111718. doi:10.1016/j.jphotobiol.2019.111718
156. Peña-Bahamonde J, Nguyen HN, Fanourakis SK, Rodrigues DF. Recent advances in graphene-based biosensor technology with applications in life sciences. J Nanobiotechnol. 2018;16(1):75. doi:10.1186/s12951-018-0400-z
157. Ilves M, Palomäki J, Vippola M, Lehto M, Savolainen K, Savinko T, Alenius H. Topically applied ZnO nanoparticles suppress allergen induced skin inflammation but induce vigorous IgE production in the atopic dermatitis mouse model. Part Fibre Toxicol. 2014;11:38. doi:10.1186/s12989-014-0038-4
158. Saratale RG, Karuppusamy I, Saratale GD, Pugazhendhi A, Kumar G, Park Y, Ghodake GS, Bharagava RN, Banu JR, Shin HS. A comprehensive review on green nanomaterials using biological systems: recent perception and their future applications. Colloids Surf B Biointerfaces. 2018;170:20–35. doi:10.1016/j.colsurfb.2018.05.045
159. Ali A, Phull AR, Zia M. Elemental zinc to zinc nanoparticles: is ZnO NPs crucial for life? Synthesis, toxicological, and environmental concerns. Nanotechnol Rev. 2018;7(5):413–441. doi:10.1515/ntrev-2018-0067
160. Rao MD, Gautam P. Synthesis and characterization of ZnO nanoflowers using Chlamydomonas reinhardtii: a green approach. Environ Prog Sustain Energy. 2016;35(4):1020–1026. doi:10.1002/ep.12315
161. Azizi S, Ahmad MB, Namvar F, Mohamad R. Green biosynthesis and characterization of zinc oxide nanoparticles using brown marine macroalga Sargassum muticum aqueous extract. Mater Lett. 2014;116:275–277. doi:10.1016/j.matlet.2013.11.038
162. Francavilla M, Pineda A, Romero AA, Colmenares JC, Vargas C, Monteleone M, Luque R. Efficient and simple reactive milling preparation of photocatalytically active porous ZnO nanostructures using biomass derived polysaccharides. Green Chem. 2014;16(5):2876–2885. doi:10.1039/C3GC42554A
163. Nagarajan S, Arumugam Kuppusamy K. Extracellular synthesis of zinc oxide nanoparticle using seaweeds of gulf of Mannar, India. J Nanobiotechnol. 2013;11:39. doi:10.1186/1477-3155-11-39
164. Murugan K, Roni M, Panneerselvam C, Suresh U, Rajaganesh R, Aruliah R, Mahyoub JA, Trivedi S, Rehman H, Al-Aoh HA, Kumar S. Sargassum wightii-synthesized ZnO nanoparticles reduce the fitness and reproduction of the malaria vector Anopheles stephensi and cotton bollworm Helicoverpa armigera. Physiol Mol Plant Pathol. 2018;101:202–213. doi:10.1016/j.pmpp.2017.02.004
165. Alsaggaf MS, Diab AM, ElSaied BEF, Tayel AA, Moussa SH. Application of ZnO nanoparticles phycosynthesized with Ulva fasciata extract for preserving peeled shrimp quality. Nanomaterials. 2021;11(2):385. doi:10.3390/nano11020385
166. Priyadharshini RI, Prasannaraj G, Geetha N, Venkatachalam P. Microwave-mediated extracellular synthesis of metallic silver and zinc oxide nanoparticles using macro-algae (Gracilaria edulis) extracts and its anticancer activity against human PC3 cell lines. Appl Biochem Biotechnol. 2014;174(8):2777–2790. doi:10.1007/s12010-014-1225-3
167. Thema FT, Manikandan E, Dhlamini MS, Maaza MJ. Green synthesis of ZnO nanoparticles via Agathosma betulina natural extract. Mater Lett. 2015;161:124–127. doi:10.1016/j.matlet.2015.08.052
168. Bhattacharya P, Chatterjee K, Swarnakar S, Banerjee S. Green synthesis of zinc oxide nanoparticles via algal route and its action on cancerous cells and pathogenic microbes. Adv Nano Res. 2020;3(1):15–27. doi:10.21467/anr.3.1.15-27
169. Shokoofeh N, Moradi-Shoeili Z, Naeemi AS, Jalali A, Hedayati M, Salehzadeh A. Biosynthesis of Fe3O4@ Ag nanocomposite and evaluation of its performance on expression of norA and norB efflux pump genes in ciprofloxacin-resistant Staphylococcus aureus. Biol Trace Elem Res. 2019;191(2):522–530. doi:10.1007/s12011-019-1632-y
170. Subramanian H, Krishnan M, Mahalingam A. Photocatalytic dye degradation and photoexcited anti-microbial activities of green zinc oxide nanoparticles synthesized via Sargassum muticum extracts. RSC Adv. 2022;12(2):985–997. doi:10.1039/d1ra08196a
171. Khilji SA, Munir N, Aziz I, Anwar B, Hasnain M, Jakhar AM, Sajid ZA, Abideen Z, Hussain MI, El-Habeeb AA, Yang HH. Application of algal nanotechnology for leather wastewater treatment and heavy metal removal efficiency. Sustainability. 2022;14(21):13940. doi:10.3390/su142113940
172. Osman AI, Zhang Y, Farghali M, Rashwan AK, Eltaweil AS, Abd El-Monaem EM, Mohamed, IM, Badr MM, Ihara I, Rooney DW, Yap PS. Synthesis of green nanoparticles for energy, biomedical, environmental, agricultural, and food applications: a review. Environ Chem Lett. 2024;22(2):841–887. doi:10.1007/s10311-023-01682-3
173. Parashar M, Shukla VK, Singh R. Metal oxides nanoparticles via sol–gel method: a review on synthesis, characterization and applications. J Mater Sci. 2020;31(5):3729–3749. doi:10.1007/s10854-020-02994-8
174. Wang N,Fuh JYH,Dheen ST,Senthil Kumar A. Synthesis methods of functionalized nanoparticles: a review. Bio-des Manuf. 2021;4(2):379–404. doi:10.1007/s42242-020-00106-3
175. Navas D, Ibañez A, González I, Palma JL, Dreyse P. Controlled dispersion of ZnO nanoparticles produced by basic precipitation in solvothermal processes. Heliyon. 2020;6(12):e05821. doi:10.1016/j.heliyon.2020.e05821
176. Liu W, Wang S, Wang J, Zhang B, Liu L, Liu H, Yang J. Supercritical hydrothermal synthesis of nano-zinc oxide: process and mechanism. Ceram Int. 2022;48(16):22629–22646. doi:10.1016/j.ceramint.2022.05.094
177. Izzi M, Sportelli MC, Torsi L, Picca RA, Cioffi N. Synthesis and antimicrobial applications of ZnO nanostructures: a review. ACS Appl Nano Mater. 2023;6(13):10881–10902. doi:10.1021/acsanm.3c01432
178. Bandeira M, Giovanela M, Roesch-Ely M, Devine DM, da Silva Crespo J. Green synthesis of zinc oxide nanoparticles: a review of the synthesis methodology and mechanism of formation. Sustain Chem Pharm. 2020;15:100223. doi:10.1016/j.scp.2020.100223
179. García-Quintero A, Palencia M. A critical analysis of environmental sustainability metrics applied to green synthesis of nanomaterials and the assessment of environmental risks associated with the nanotechnology. Sci Total Environ. 2021;793:148524. doi:10.1016/j.scitotenv.2021.148524
180. Kumar JA, Krithiga T, Manigandan S, Sathish S, Renita AA, Prakash P, Prasad BN, Kumar TP, Rajasimman M, Hosseini-Bandegharaei A, Prabu D. A focus to green synthesis of metal/metal based oxide nanoparticles: various mechanisms and applications towards ecological approach. J Clean Prod. 2021;324:129198. doi:10.1016/j.jclepro.2021.129198
181. Al-darwesh MY, Ibrahim SS, Mohammed MA. A review on plant extract mediated green synthesis of zinc oxide nanoparticles and their biomedical applications. Results Chem. 2024;7:101368. doi:10.1016/j.rechem.2024.101368
182. Chehelgerdi M, Chehelgerdi M, Allela OQB, Pecho RDC, Jayasankar N, Rao DP, Thamaraikani T, Vasanthan M, Viktor P, Lakshmaiya N, Saadh MJ, Amajd A, Abo-Zaid MA, Castillo-Acobo RY, Ismail AH, Amin AH, Akhavan-Sigari R. Progressing nanotechnology to improve targeted cancer treatment: overcoming hurdles in its clinical implementation. Mol Cancer. 2023;22(1):169. doi:10.1186/s12943-023-01865-0
183. Nie C, Ma L, Li S, Fan X, Yang Y, Cheng C, Zhao W, Zhao C. Recent progresses in graphene-based bio-functional nanostructures for advanced biological and cellular interfaces. Nano Today. 2019;26:57–97. doi:10.1016/j.nantod.2019.03.003
184. Husen A. Natural product-based fabrication of zinc-oxide nanoparticles and their applications. In: Husen A, Iqbal M., editors. Nanomaterials and Plant Potential. Cham. Switzerland: Springer; 2019:193–219. doi:10.1007/978-3-030-05569-1_7
185. Mohamad Sukri SNA, Shameli K, Teow SY, Chew J, Ooi LT, Lee-Kiun Soon M, Ismail NA, Moeini H. Enhanced antibacterial and anticancer activities of plant extract mediated green synthesized zinc oxide-silver nanoparticles. Front Microbiol. 2023;14:1194292. doi:10.3389/fmicb.2023.1194292
186. Naiel B, Fawzy M, Halmy MWA, Mahmoud AED. Green synthesis of zinc oxide nanoparticles using sea lavender (Limonium pruinosum L. Chaz.) extract: characterization, evaluation of anti-skin cancer, antimicrobial and antioxidant potentials. Sci Rep. 2022;12(1):20370. doi:10.1038/s41598-022-24805-2
187. Sanaeimehr Z, Javadi I, Namvar F. Antiangiogenic and antiapoptotic effects of green-synthesized zinc oxide nanoparticles using Sargassum muticum algae extraction. Cancer Nanotechnol. 2018;9(1):3. doi:10.1186/s12645-018-0037-5
188. Tabrez S, Khan AU, Hoque M, Suhail M, Khan MI, Zughaibi TA. Biosynthesis of ZnO NPs from pumpkin seeds’ extract and elucidation of its anticancer potential against breast cancer. Nanotechnol Rev. 2022;11(1):2714–2725. doi:10.1515/ntrev-2022-0154
189. Karimi N, Behbahani M, Dini G, Razmjou A. Anticancer effects of Echinacea purpurea extracts, treated with green synthesized ZnO nanoparticles on human breast cancer (MCF-7) and PBMCs proliferation. Mater Res Express. 2019;6(9):095402. doi:10.1088/2053-1591/ab29d2
190. Sarala E, Madhukara Naik M, Vinuth M, Rami Reddy YV, Sujatha HR. Green synthesis of Lawsonia inermis-mediated zinc ferrite nanoparticles for magnetic studies and anticancer activity against breast cancer (MCF-7) cell lines. J Mater Sci Mater Electron. 2020;31:8589–8596. doi:10.1007/s10854-020-03394-8
191. Cheng J, Wang X, Qiu L, Li Y, Marraiki N, Elgorban AM, Xue L. Green synthesized zinc oxide nanoparticles regulates the apoptotic expression in bone cancer cells MG-63 cells. J Photochem Photobiol B. 2020;202:111644. doi:10.1016/j.jphotobiol.2019.111644
192. Efati Z, Shahangian SS, Darroudi M, Amiri H, Hashemy SI, Aghamaali MR. Green chemistry synthesized zinc oxide nanoparticles in Lepidium sativum L. seed extract and evaluation of their anticancer activity in human colorectal cancer cells. Ceram Int. 2023;49(20):32568–32576. doi:10.1016/j.ceramint.2023.07.221
193. Jobie FN, Ranjbar M, Moghaddam AH, Kiani M. Green synthesis of zinc oxide nanoparticles using Amygdalus scoparia Spach stem bark extract and their applications as an alternative antimicrobial, anticancer, and anti-diabetic agent. Adv Powder Technol. 2021;32(6):2043–2052. doi:10.1016/j.apt.2021.04.014
194. Selim YA, Azb MA, Ragab I H MAbd El-Azim M. Green synthesis of zinc oxide nanoparticles using aqueous extract of Deverra tortuosa and their cytotoxic activities. Sci Rep. 2020;10(1):3445. doi:10.1038/s41598-020-60541-1
195. Vijayakumar S, Vaseeharan B, Malaikozhundan B, Shobiya M. Laurus nobilis leaf extract mediated green synthesis of ZnO nanoparticles: characterization and biomedical applications. Biomed Pharmacother. 2016;84:1213–1222. doi:10.1016/j.biopha.2016.10.038
196. Chung IM, Rahuman AA, Marimuthu S, Kirthi AV, Anbarasan K, Rajakumar G. An investigation of the cytotoxicity and caspase-mediated apoptotic effect of green synthesized zinc oxide nanoparticles using Eclipta prostrata on human liver carcinoma cells. Nanomaterials. 2015;5(3):1317–1330. doi:10.3390/nano5031317
197. Mohammed YHI, Alghamdi S, Jabbar B, Marghani D, Beigh S, Abouzied AS, Khalifa NE, Khojali WMA, Huwaimel B, Alkhalifah DHM, Hozzein WN. Green synthesis of zinc oxide nanoparticles using Cymbopogon citratus extract and its antibacterial activity. ACS Omega. 2023;8(35):32027–32042. doi:10.1021/acsomega.3c03908
198. Imade EE, Ajiboye TO, Fadiji AE, Onwudiwe DC, Babalola OO. Green synthesis of zinc oxide nanoparticles using plantain peel extracts and the evaluation of their antibacterial activity. Sci Afr. 2022;
199. Abdelsattar AS, Kamel AG, Hussein AH, Azzam M, Makky S, Rezk N, Essam K, Agwa MM, El-Shibiny A. The promising antibacterial and anticancer activity of green synthesized zinc nanoparticles in combination with silver and gold nanoparticles. J Inorg Organomet Polym. 2023;33(7):1868–1881. doi:10.1007/s10904-023-02614-y
200. Iqbal J, Abbasi BA, Yaseen T, Zahra SA, Shahbaz A, Shah SA, Uddin S, Ma X, Raouf B, Kanwal S, Amin W, Mahmood T, El-Serehy HA, Ahmad P. Green synthesis of zinc oxide nanoparticles using Elaeagnus angustifolia L. leaf extracts and their multiple in vitro biological applications. Sci Rep. 2021;11(1):20988. doi:10.1038/s41598-021-99839-z
201. Happy A, Soumya M, Venkat Kumar S, Rajeshkumar S, Sheba RD, Lakshmi T, Deepak Nallaswamy V. Phyto-assisted synthesis of zinc oxide nanoparticles using Cassia alata and its antibacterial activity against Escherichia coli. Biochem Biophys Rep. 2019;17:208–211. doi:10.1016/j.bbrep.2019.01.002
202. Anbuvannan M, Ramesh M, Viruthagiri G, Shanmugam N, Kannadasan N. Synthesis, characterization and photocatalytic activity of ZnO nanoparticles prepared by biological method. Spectrochim Acta A. 2015;143:304–308. doi:10.1016/j.saa.2015.01.124
203. Ramesh M, Anbuvannan M, Viruthagiri G. Green synthesis of ZnO nanoparticles using Solanum nigrum leaf extract and their antibacterial activity. Spectrochim Acta A. 2015;136:864–870. doi:10.1016/j.saa.2014.09.105
204. Ramesh A, Sundarraj P, Balamani J. A potent cytotoxicity and antimicrobial activity of zinc oxide nanoparticles synthesized by leaf of Ipomoea Pes-Caprae (L.) R. BR. Asian J Pharm Clin Res. 2019;12(5):111–117. doi:10.22159/ajpcr.2019.v12i5.32318
205. Gupta M, Tomar RS, Kaushik S, Mishra RK, Sharma D. Effective antimicrobial activity of green ZnO nanoparticles of Catharanthus roseus. Front Microbiol. 2018;9:2030. doi:10.3389/fmicb.2018.02030
206. Dobrucka R, Długaszewska J. Biosynthesis and antibacterial activity of ZnO nanoparticles using Trifolium pratense flower extract. Saudi J Biol Sci. 2016;23(4):517–523. doi:10.1016/j.sjbs.2015.05.016
207. Ahmed S, Annu, Chaudhry SA, Ikram S. A review on biogenic synthesis of ZnO nanoparticles using plant extracts and microbes: a prospect towards green chemistry. J Photoch Photobiol B. 2017;166:272–284. doi:10.1016/j.jphotobiol.2016.12.011
208. Murali M, Mahendra C, Nagabhushan Rajashekar, N, Sudarshana MSRaveesha KA, Amruthesh KN. Antibacterial and antioxidant properties of biosynthesized zinc oxide nanoparticles from Ceropegia candelabrum L. an endemic species. Spectrochim Acta A. 2017;179:104–109. doi:10.1016/j.saa.2017.02.027
209. Kalpana VN, Devi Rajeswari V. A review on green synthesis, biomedical applications, and toxicity studies of ZnO NPs. Bioinorg Chem Appl. 2018;2018(1):3569758. doi:10.1155/2018/3569758
210. Sathishkumar G, Rajkuberan C, Manikandan K, Prabukumar S, DanielJohn J, Sivaramakrishnan S. Facile biosynthesis of antimicrobial zinc oxide (ZnO) nanoflakes using leaf extract of Couroupita guianensis. Aubl Mater Lett. 2017;188:383–386. doi:10.1016/j.matlet.2016.11.100
211. Rajiv P, Rajeshwari S, Venckatesh R. Bio-Fabrication of zinc oxide nanoparticles using leaf extract of Parthenium hysterophorus L. and its size-dependent antifungal activity against plant fungal pathogens. Spectrochim Acta A. 2013;112:384–387. doi:10.1016/j.saa.2013.04.072
212. Hameed H, Waheed A, Sharif MS, Saleem M, Afreen A, Tariq M, Kamal A, Al-Onazi WA, Al Farraj DA, Ahmad S, Mahmoud RM. Green synthesis of zinc oxide (Zno) nanoparticles from green algae and their assessment in various biological applications. Micromachines. 2023;14(5):928. doi:10.3390/mi14050928
213. Agarwal H, Shanmugam V. A review on anti-inflammatory activity of green synthesized zinc oxide nanoparticle: mechanism-based approach. Bioorg Chem. 2020;94:103423. doi:10.1016/j.bioorg.2019.103423
214. Rajakumar G, Thiruvengadam M, Mydhili G, Gomathi T, Chung IM. Green approach for synthesis of zinc oxide nanoparticles from Andrographis paniculata leaf extract and evaluation of their antioxidant, anti-diabetic, and anti-inflammatory activities. Bioprocess Biosyst Eng. 2018;41(1):21–30. doi:10.1007/s00449-017-1840-9
215. Nagajyothi PC, Cha SJ, Yang IJ, Sreekanth TV, Kim KJ, Shin HM. Antioxidant and anti-inflammatory activities of zinc oxide nanoparticles synthesized using Polygala tenuifolia root extract. J Photochem Photobiol B. 2015;146:10–17. doi:10.1016/j.jphotobiol.2015.02.008
216. Mohammad GRKS, Tabrizi MH, Ardalan T, Yadamani S, Safavi E. Green synthesis of zinc oxide nanoparticles and evaluation of anti-angiogenesis, anti-inflammatory and cytotoxicity properties. J Biosci. 2019;44(2):30. doi:10.1007/s12038-019-9845-y
217. Agarwal H, Shanmugam VK. Synthesis and optimization of zinc oxide nanoparticles using Kalanchoe pinnata towards the evaluation of its anti-inflammatory activity. J Drug Deliv Sci Technol. 2019;54:101291. doi:10.1016/j.jddst.2019.101291
218. Abdelbaky AS, Abd El-Mageed TA, Babalghith AO, Selim S, Mohamed AMHA. Green synthesis and characterization of ZnO nanoparticles using Pelargonium odoratissimum (L.) aqueous leaf extract and their antioxidant, antibacterial and anti-inflammatory activities. Antioxidants. 2022;11(8):1444. doi:10.3390/antiox11081444
219. Manasa DJ, Chandrashekar KR, Kumar MP, Suresh D, Kumar DM, Ravikumar CR, Bhattacharya T, Murthy HA. Proficient synthesis of zinc oxide nanoparticles from Tabernaemontana heyneana Wall. via green combustion method: antioxidant, anti-inflammatory, antidiabetic, anticancer and photocatalytic activities. Results Chem. 2021;3:100178. doi:10.1016/j.rechem.2021.100178
220. Thatoi P, Kerry RG, Gouda S, Das G, Pramanik K, Thatoi H, Patra JK. Photo-mediated green synthesis of silver and zinc oxide nanoparticles using aqueous extracts of two mangrove plant species, Heritiera fomes and Sonneratia apetala and investigation of their biomedical applications. J Photochem Photobiol B. 2016;163:311–318. doi:10.1016/j.jphotobiol.2016.07.029
221. Diab T, Alkafaas SS, Shalaby TI, Hessien M. Paclitaxel nanoparticles induce apoptosis and regulate TXR1, CYP3A4 and CYP2C8 in breast cancer and hepatoma cells. Anticancer Agents Med Chem. 2020;20(13):1582–1591. doi:10.2174/1871520620666200504071530
222. Alkafaas SS, Diab T, Shalaby T, Hessien M. Dexamethasone improves the responsiveness of hepatoma cells for both free and solvent containing paclitaxel in vitro. Egypt J Biochem Mol Biol. 2019;37(1–2):95–110.
223. Elsalahaty MI, Alkafaas SS, Bashir AO, El-Tarabily KA, El-Saadony MT, Yousef EH. Revealing the association between vitamin D metabolic pathway gene variants and lung cancer risk: a systematic review and meta-analysis. Front Genet. 2024;15:1302527. doi:10.3389/fgene.2024.1302527
224. Martínez-Carmona M, Gun’ko Y, Vallet-Regí M. ZnO Nanostructures for drug delivery and theranostic applications. Nanomaterials. 2018;8(4):268. doi:10.3390/nano8040268
225. Wiesmann N, Tremel W, Brieger J. Zinc oxide nanoparticles for therapeutic purposes in cancer medicine. J Mater Chem B. 2020;8(23):4973–4989. doi:10.1039/D0TB00739K
226. Siddiqi KS, Ur Rahman A, Tajuddin N, Husen A. Properties of zinc oxide nanoparticles and their activity against microbes. Nanoscale Res Lett. 2018;13(1):141. doi:10.1186/s11671-018-2532-3
227. Al-darwesh MY, Babakr KA, Qader IN. Characterization and anticancer evaluation of zirconia nanoparticles synthesized via green route using Sophora flavescens roots extract. Nano-Struct Nano-Objects. 2024;39:101245. doi:10.1016/j.nanoso.2024.101245
228. Omar MF, Aziz HA, Stoll S. Stability of ZnO nanoparticles in solution. Influence of pH, dissolution, aggregation and disaggregation effects. J Colloid Sci Biotechnol. 2014;3(1):75–84. doi:10.1166/jcsb.2014.1072
229. Tate PM, Mastrodomenico V, Cunha C, McClure J, Barron AE, Diamond G, Mounce BC, Kirshenbaum K. Peptidomimetic oligomers targeting membrane phosphatidylserine exhibit broad antiviral activity. ACS Infect Dis. 2023;9(8):1508–1522. doi:10.1021/acsinfecdis.3c00063
230. Taylor Z, Marucho M. The self-adaptation ability of zinc oxide nanoparticles enables reliable cancer treatments. Nanomaterials. 2020;10(2):269. doi:10.3390/nano10020269
231. Singh S, Jaiswal V, Singh JK, Semwal R, Raina D. Nanoparticle formulations: a smart era of advanced treatment with nanotoxicological imprints on the human body. Chem Biol Interact. 2023;373:110355. doi:10.1016/j.cbi.2023.110355
232. Youssef FS, Ismail SH, Fouad OA, Mohamed GG. Green synthesis and biomedical applications of zinc oxide nanoparticles. Review J Egypt Vet Med. 2024;55(1):287–311. doi:10.21608/ejvs.2023.225862.1576
233. Chandrasekaran S, Anusuya S, Anbazhagan V. Anticancer, anti-diabetic, antimicrobial activity of zinc oxide nanoparticles: a comparative analysis. J Mol Struct. 2022;1263:133139. doi:10.1016/j.molstruc.2022.133139
234. Raj S, Khurana S, Choudhari R, Kesari KK, Kamal MA, Garg N, Ruokolainen J, Das BC, Kumar D. Specific targeting cancer cells with nanoparticles and drug delivery in cancer therapy. Semin Cancer Biol. 2021;69:166–177. doi:10.1016/j.semcancer.2019.11.002
235. Ghaffari SB, Sarrafzadeh MH, Fakhroueian Z, Shahriari S, Khorramizadeh MR. Functionalization of ZnO nanoparticles by 3-mercaptopropionic acid for aqueous curcumin delivery: synthesis, characterization, and anticancer assessment. Mater Sci Eng C Mater Biol Appl. 2017;79:465–472. doi:10.1016/j.msec.2017.05.065
236. Zhang J, Qin X, Wang B, Xu G, Qin Z, Wang J, Wu L, Ju X, Bose DD, Qiu F, Zhou H. Zinc oxide nanoparticles harness autophagy to induce cell death in lung epithelial cells. Cell Death Dis. 2017;8(7):e2954. doi:10.1038/cddis.2017.337
237. Batool M, Khurshid S, Daoush WM, Siddique SA, Nadeem T. Green synthesis and biomedical applications of ZnO nanoparticles: role of PEGylated-ZnO nanoparticles as doxorubicin drug carrier against MDA-MB-231 (TNBC) cells line. Crystals. 2021;11(4):344. doi:10.3390/cryst11040344
238. Gomathi R, Suhana H, Paradesi D. Characterization study of cytotoxicity of green synthesized ZnO nanoparticles loaded with anti‐cancer doxorubicin drug. Chem Select. 2021;6(18):4533–4538. doi:10.1002/slct.202100358
239. da Silva BL, Abuçafy MP, Berbel Manaia E, Oshiro Junior JA, Chiari-Andréo BG, Pietro RCR, Chiavacci LA. Relationship between structure and antimicrobial activity of zinc oxide nanoparticles: an overview. Int J Nanomed. 2019;14:9395–9410. doi:10.2147/IJN.S216204
240. Jin SE, Jin HE. Synthesis, characterization, and three-dimensional structure generation of zinc oxide-based nanomedicine for biomedical applications. Pharmaceutics. 2019;11(11):575. doi:10.3390/pharmaceutics11110575
241. Sudhakaran S, Athira SS, Mohanan PV. Zinc oxide nanoparticle induced neurotoxic potential upon interaction with primary astrocytes. Neurotoxicology. 2019;73:213–227. doi:10.1016/j.neuro.2019.04.008
242. Ali F, Neha K, Parveen S. Current regulatory landscape of nanomaterials and nanomedicines: a global perspective. J Drug Deliv Technol. 2023;80:104118. doi:10.1016/j.jddst.2022.104118
243. Díez-Pascual AM. Surface engineering of nanomaterials with polymers, biomolecules, and small ligands for nanomedicine. Materials. 2022;15(9):3251. doi:10.3390/ma15093251
244. Yang T, Zhai J, Hu D, Yang R, Wang G, Li Y, Liang G. “Targeting design” of nanoparticles in tumor therapy. Pharmaceutics. 2022;14(9):1919. doi:10.3390/pharmaceutics14091919
245. Ramana V, Rajeshkumar S, Jagadeesh K. Review of the environmentally friendly production of zinc oxide nanoparticles and its anti-oxidant, anti-hyperlipidemic, and anti-diabetic properties. J Surv Fish Sci. 2023;10(1S):117–127. doi:10.17762/sfs.v10i1S.154
246. Gadoa ZA, Moustafa AH, El Rayes SM, Arisha AA, Mansour MF. Zinc Oxide nanoparticles and synthesized pyrazolopyrimidine alleviate diabetic effects in rats induced by Type II diabetes. ACS Omega. 2022;7(41):36865–36872. doi:10.1021/acsomega.2c05638
247. Jiang J, Pi J, Cai J. The advancing of zinc oxide nanoparticles for biomedical applications. Bioinorg Chem. 2018;2018(1):1062562. doi:10.1155/2018/1062562
248. Li X, Bai Y, Jin Z, Svensson B. Food-derived non-phenolic α-amylase and α-glucosidase inhibitors for controlling starch digestion rate and guiding diabetes-friendly recipes. LWT. 2022;153:112455. doi:10.1016/j.lwt.2021.112455
249. Rehana D, Mahendiran D, Kumar RS, Rahiman AK. In vitro antioxidant and antidiabetic activities of zinc oxide nanoparticles synthesized using different plant extracts. Bioprocess Biosyst Eng. 2017;40(6):943–957. doi:10.1007/s00449-017-1758-2
250. Vinotha V, Iswarya A, Thaya R, Govindarajan M, Alharbi NS, Kadaikunnan S, Khaled JM, Al-Anbr MN, Vaseeharan B. Synthesis of ZnO nanoparticles using insulin-rich leaf extract: anti-diabetic, antibiofilm and anti-oxidant properties. J Photochem Photobiol B. 2019;197:111541. doi:10.1016/j.jphotobiol.2019.111541
251. Lin Q, Qiu C, Li X, Sang S, McClements DJ, Chen L, Long J, Jiao A, Tian Y, Jin Z. The inhibitory mechanism of amylase inhibitors and research progress in nanoparticle-based inhibitors. Crit Rev Food Sci Nutr. 2023;63(33):12126–12135. doi:10.1080/10408398.2022.2098687
252. Mittal A, Gandhi S, Roy I. Mechanistic interaction studies of synthesized ZIF-8 nanoparticles with bovine serum albumin using spectroscopic and molecular docking approaches. Sci Rep. 2022;12(1):10331. doi:10.1038/s41598-022-14630-y
253. Udnoor A, Lokolkar M, Yallur BC, Kale R, Kalasad MN, Katrahalli U, Manjunatha DH. Monitoring the interactions between bovine serum albumin and ZnO/Ag nanoparticles by spectroscopic techniques. J Biomol Struct Dyn. 2023;41(1):352–365. doi:10.1080/07391102.2021.2006788
254. Nazarizadeh A, Asri-Rezaie S. Comparative study of antidiabetic activity and oxidative stress induced by zinc oxide nanoparticles and zinc sulfate in diabetic rats. AAPS Pharm Sci Tech. 2016;17(4):834–843. doi:10.1016/j.jphotobiol.2019.111541
255. Mukhtar Y, Galalain A, Yunusa U. A modern overview on Diabetes mellitus: a chronic endocrine disorder. Eur J Biol. 2020;5(2):1–14. doi:10.47672/ejb.409
256. Zhao T, Huang Q, Su Y, Sun W, Huang Q, Wei W. Zinc and its regulators in pancreas. Inflammopharmacology. 2019;27:453–464. doi:10.1007/s10787-019-00573-w
257. Shoaib A, Shahid S, Mansoor S, Javed M, Iqbal S, Mahmood S, Bahadur A, Jaber F, Alshalwi M. Tailoring of an anti-diabetic drug empagliflozin onto zinc oxide nanoparticles: characterization and in vitro evaluation of anti-hyperglycemic potential. Sci Rep. 2024;14(1):2499. doi:10.1038/s41598-024-52523-4
258. El-Daly SM, Medhat DA El-Bana M, Abdel-Latif Y, El-Naggar ME, Omara EA, Morsy SM, Hussein J. Stimulatory effect of docosahexaenoic acid alone or loaded in zinc oxide or silver nanoparticles on the expression of glucose transport pathway. Prostaglandins Other Lipid Mediat. 2021;155:106566. doi:10.1016/j.prostaglandins.2021.106566
259. Ruan S, Guo X, Ren Y, Cao G, Xing H, Zhang X. Nanomedicines based on trace elements for intervention of Diabetes mellitus. Biomed Pharmacother. 2023;168:115684. doi:10.1016/j.biopha.2023.115684
260. Eswari KM, Asaithambi S, Karuppaiah M, Sakthivel P, Balaji V, Ponelakkia DK, Yuvakkumar R, Kumar P, Vijayaprabhu N, Ravi G. Green synthesis of ZnO nanoparticles using Abutilon Indicum and Tectona grandis leaf extracts for evaluation of anti-diabetic, anti-inflammatory and in vitro cytotoxicity activities. Ceram Int. 2022;48(22):33624–33634. doi:10.1016/j.ceramint.2022.07.308
261. Ahluwalia KK, Thakur K, Ahluwalia AS, Hashem A, Avila-Quezada GD, Abd Allah EF, Thakur N. Assessment of genotoxicity of zinc oxide nanoparticles using mosquito as test model. Toxics. 2023;11(11):887. doi:10.3390/toxics11110887
262. Allan J, Belz S, Hoeveler A, Hugas M, Okuda H, Patri A, Rauscher H, Silva P, Slikker W, Sokull-Kluettgen B. Regulatory landscape of nanotechnology and nanoplastics from a global perspective. Regul Toxicol Pharmacol. 2021;122:104885. doi:10.1016/j.yrtph.2021.104885
263. Virgen-Ortiz A, Apolinar-Iribe A, Díaz-Reval I, Parra-Delgado H, Limón-Miranda S, Sánchez-Pastor EA, Castro-Sánchez L, Jesús Castillo S, Dagnino-Acosta A, Bonales-Alatorre E, Rodríguez-Hernández A. Zinc oxide nanoparticles induce an adverse effect on blood glucose levels depending on the dose and route of administration in healthy and diabetic rats. Nanomaterials. 2020;10(10):2005. doi:10.3390/nano10102005
264. Stoleru OA, Burlec AF, Mircea C, Felea MG, Macovei I, Hăncianu M, Corciovă A. Multiple nanotechnological approaches using natural compounds for diabetes management. J Diabetes Metab Disord. 2024;23:267–287. doi:10.1007/s40200-023-01376-1
265. Fadwa AO, Alkoblan DK, Mateen A, Albarag AM. Synergistic effects of zinc oxide nanoparticles and various antibiotics combination against Pseudomonas aeruginosa clinically isolated bacterial strains. Saudi J Biol Sci. 2021;28(1):928–935. doi:10.1016/j.sjbs.2020.09.064
266. Helmy YA, Taha-Abdelaziz K, Hawwas HAE, Ghosh S, AlKafaas SS, Moawad MMM, Saied EM, Kassem II, Mawad AMM. Antimicrobial resistance and recent alternatives to antibiotics for the control of bacterial pathogens with an emphasis on foodborne pathogens. Antibiotics. 2023;12(2):274. doi:10.3390/antibiotics12020274
267. Alduhaidhawi AHM, AlHuchaimi SN, Al-Mayah TA, Al-Ouqaili MTS, Alkafaas SS, Muthupandian S, Saki M. Prevalence of CRISPR-Cas Systems and their possible association with antibiotic resistance in Enterococcus faecalis and Enterococcus faecium collected from hospital wastewater. Infect Drug Resist. 2022;15:1143–1154. doi:10.2147/IDR.S358248
268. Akinyemi KO, Al-Khafaji NSK, Al-Alaq FT, Fakorede CO, Al-Dahmoshi HOM, Iwalokun BA, Akpabio I, Alkafaas SS, Saki M. Extended-spectrum Beta-lactamases encoding genes among Salmonella enterica serovar Typhi isolates in patients with typhoid fever from four academic medical centers Lagos, Nigeria. Rev Invest Clin. 2022;74(3):165–171. doi:10.24875/RIC.22000078
269. Wang L, Hu C, Shao L. The antimicrobial activity of nanoparticles: present situation and prospects for the future. Int J Nanomed. 2017;12:1227–1249. doi:10.2147/IJN.S121956
270. Al-Momani H, Al Balawi D, Hamed S, Albiss BA, Almasri M, AlGhawrie H, Ibrahim L, Al Balawi H, Al Haj Mahmoud S, Pearson J, Ward C. The impact of biosynthesized ZnO nanoparticles from Olea europaea (Common olive) on Pseudomonas aeruginosa growth and biofilm formation. Sci Rep. 2023;13(1):5096. doi:10.1038/s41598-023-32366-1
271. Doan Thi TU, Nguyen TT, Thi YD, Ta Thi KH, Phan BT, Pham KN. Green synthesis of ZnO nanoparticles using orange fruit peel extract for antibacterial activities. RSC Adv. 2020;10(40):23899–23907. doi:10.1039/d0ra04926c
272. Núño K. Antimicrobial Activity of Zinc Oxide Nanoparticles Compared to Silver Nanoparticles Administered Towards methicillin-Resistant Staphylococcus Aureus Biofilms. University of South Florida ProQuest Dissertations Publishing; 2022:29061234.
273. Amicizia D, Micale RT, Pennati BM, Zangrillo F, Iovine M, Lecini E, Marchini F, Lai PL, Panatto D. Burden of typhoid fever and cholera: similarities and differences. Prevention strategies for European travelers to endemic/epidemic areas. J Prev Med Hyg. 2019;60(4):E271–E285. doi:10.15167/2421-4248/jpmh2019.60.4.1333
274. Sarwar S, Chakraborti S, Bera S, Sheikh IA, Hoque KM, Chakrabarti P. The antimicrobial activity of ZnO nanoparticles against Vibrio cholerae: variation in response depends on biotype. Nanomedicine. 2016;12(6):1499–1509. doi:10.1016/j.nano.2016.02.006
275. Sarwar S, Ali A, Pal M, Chakrabarti P. Zinc oxide nanoparticles provide anti-cholera activity by disrupting the interaction of cholera toxin with the human GM1 receptor. J Biol Chem. 2017;292(44):18303–18311. doi:10.1074/jbc.M117.793240
276. Augustine R, Hasan A, Primavera R, Wilson RJ, Thakor AS, Kevadiya BD. Cellular uptake and retention of nanoparticles: insights on particle properties and interaction with cellular components. Mater Today Commun. 2020;25:101692. doi:10.1016/j.mtcomm.2020.101692
277. Madkour LH. Reactive Oxygen Species (ROS), Nanoparticles, and Endoplasmic Reticulum (ER) Stress-Induced Cell Death Mechanisms. Cambridge, Massachusetts, USA: AcademicPress; 2020:780.
278. Mohammadinejad R, Moosavi MA, Tavakol S, Vardar DÖ, Hosseini A, Rahmati M, Dini L, Hussain S, Mandegary A, Klionsky DJ. Necrotic, apoptotic and autophagic cell fates triggered by nanoparticles. Autophagy. 2019;15(1):4–33. doi:10.1080/15548627.2018.1509171
279. Summer M, Ashraf R, Ali S, Bach H, Noor S, Noor Q, Riaz S, Khan RRM. Inflammatory response of nanoparticles: mechanisms, consequences, and strategies for mitigation. Chemosphere. 2024;363:142826. doi:10.1016/j.chemosphere.2024.142826
280. Heikal YM, Şuţan NA. Mechanisms of genotoxicity and oxidative stress induced by engineered nanoparticles in plants. In: Khan Z, Ansari MYK, Shahwar D., editors. Induced Genotoxicity and Oxidative Stress in Plants. Springer Singapore: Singapore; 2021:151–197. doi:10.1007/978-981-16-2074-4_6
281. Ghasemi F, Jalal R. Antimicrobial action of zinc oxide nanoparticles in combination with ciprofloxacin and ceftazidime against multidrug-resistant Acinetobacter baumannii. J Glob Antimicrob Resist. 2016;6:118–122. doi:10.1016/j.jgar.2016.04.007
282. Al-darwesh MY, Ibrahim SS, Hamid LL. Ficus carica latex mediated biosynthesis of zinc oxide nanoparticles and assessment of their antibacterial activity and biological safety. Nano-Struct Nano-Objects. 2024;38:101163. doi:10.1016/j.nanoso.2024.101163
283. Mendes CR, Dilarri G, Forsan CF, Sapata VMR, Lopes PRM, de Moraes PB, Montagnolli RN, Ferreira H, Bidoia ED. Antibacterial action and target mechanisms of zinc oxide nanoparticles against bacterial pathogens. Sci Rep. 2022;12(1):2658. doi:10.1038/s41598-022-06657-y
284. Chen J, Zhang X, Cai H, Chen Z, Wang T, Jia L, Wang J, Wan Q, Pei X. Osteogenic activity and antibacterial effect of zinc oxide/carboxylated graphene oxide nanocomposites: preparation and in vitro evaluation. Colloids Surf B Biointerfaces. 2016;147:397–407. doi:10.1016/j.colsurfb.2016.08.023
285. Sahu BP, Baishya R, Hatiboruah JL, Laloo D, Biswas N. A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. J Pharm Investig. 2022;52(5):539–585. doi:10.1007/s40005-022-00583-x
286. Javid H, Oryani MA Rezagholinejad N, Esparham ATajaldini M Karimi-Shahri M. RGD peptide in cancer targeting: benefits, challenges, solutions, and possible integrin–RGD interactions. Cancer Med. 2024;13(2):6800. doi:10.1002/cam4.6800
287. Mokhtarzadeh A, Tabarzad M, Ranjbari J, de la Guardia M, Hejazi M, Ramezani M. Aptamers as smart ligands for nano-carriers targeting. TRAC-Trends Anal Chem. 2016;82:316–327. doi:10.1016/j.trac.2016.06.018
288. Fröhlich E. The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles. Int J Nanomed. 2012;7:5577–5591. doi:10.2147/IJN.S36111
289. Zalba S, ten Hagen TLM,Burgui C,Garrido MJ. Stealth nanoparticles in oncology: facing the PEG dilemma. J Control Release. 2022;351:22–36. doi:10.1016/j.jconrel.2022.09.002
290. Mills JA, Liu F, Jarrett TR, Fletcher NL,Thurecht KJ. Nanoparticle based medicines: approaches for evading and manipulating the mononuclear phagocyte system and potential for clinical translation. Biomater Sci. 2022;10(12):3029–3053. doi:10.1039/D2BM00181K
291. Liu W-L, Zou M-Z, Qin S-Y, Cheng Y-J, Ma Y-H, Sun Y-X, Zhang X-Z. Recent advances of cell membrane-coated nanomaterials for biomedical applications. Adv Funct Mater. 2020;30(39):2003559. doi:10.1002/adfm.202003559
292. Salahpour Anarjan F. Active targeting drug delivery nanocarriers: ligands. Nano-Struct Nano-Objects. 2019;19:100370. doi:10.1016/j.nanoso.2019.100370
293. Means N, Elechalawar CK, Chen WR, Bhattacharya R, Mukherjee P. Revealing macropinocytosis using nanoparticles. Mol Asp Med. 2022;83:100993. doi:10.1016/j.mam.2021.100993
294. Barua S, Mitragotri S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: a review of current status and future prospects. Nano Today. 2014;9(2):223–243. doi:10.1016/j.nantod.2014.04.008
295. Nakamura Y, Mochida A, Choyke PL, Kobayashi H. Nanodrug delivery: is the enhanced permeability and retention effect sufficient for curing cancer? Bioconjugate Chem. 2016;27(10):2225–2238. doi:10.1021/acs.bioconjchem.6b00437
296. Subhan MA, Yalamarty SSK, Filipczak N, Parveen F, Torchilin VP. Recent advances in tumor targeting via EPR effect for cancer treatment. J Pers Med. 2021;11(6):571. doi:10.3390/jpm11060571
297. Sun L, Liu H, Ye Y, Lei Y, Islam R, Tan S, Tong R, Miao Y-B, Cai L. Smart nanoparticles for cancer therapy. Signal Transduct Target Ther. 2023;8(1):418. doi:10.1038/s41392-023-01642-x
298. Pan Y, Liu L, Mou X, Cai Y. Nanomedicine strategies in conquering and utilizing the cancer hypoxia environment. ACS Nano. 2023;17(21):20875–20924. doi:10.1021/acsnano.3c07763
299. Makabenta JM, Nabawy A, Li CH, Schmidt-Malan S, Patel R, Rotello VM. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat Rev Microbiol. 2021;19(1):23–36. doi:10.1038/s41579-020-0420-1
300. Mirzaei H, Darroudi M. Zinc oxide nanoparticles: biological synthesis and biomedical applications. Ceram Int. 2017;43(1):907–914. doi:10.1016/j.ceramint.2016.10.051
301. Islam F, Shohag S, Uddin MJ, Islam MR, Nafady MH, Akter A, Mitra S, Roy A, Emran TB, Cavalu S. Exploring the journey of zinc oxide nanoparticles (ZnO-NPs) toward biomedical applications. Materials. 2022;15(6):2160. doi:10.3390/ma15062160
302. Jin SE, Jin HE. Antimicrobial activity of zinc oxide nano/microparticles and their combinations against pathogenic microorganisms for biomedical applications: from physicochemical characteristics to pharmacological aspects. Nanomaterials. 2021;11(2). doi:10.3390/nano11020263
303. Adhikari S, Gupta R, Surin A, Kumar TS, Chakraborty S, Sarkar D, Madras G. Visible light assisted improved photocatalytic activity of combustion synthesized spongy-ZnO towards dye degradation and bacterial inactivation. RSC Adv. 2016;6(83):80086–80098. doi:10.1039/C6RA10472J
304. Kaliraj L, Ahn JC, Rupa EJ, Abid S, Lu J, Yang DC. Synthesis of panos extract mediated ZnO nano-flowers as photocatalyst for industrial dye degradation by UV illumination. J Photochem Photobiol B. 2019;199:111588. doi:10.1016/j.jphotobiol.2019.111588
305. Li Y, Liao C, Tjong SC. Recent advances in zinc oxide nanostructures with antimicrobial activities. Int J Mol Sci. 2020;21(22):8836. doi:10.3390/ijms21228836
306. Pino P, Bosco F, Mollea C, Onida B. Antimicrobial nano-zinc oxide biocomposites for wound healing applications: a review. Pharmaceutics. 2023;15(3):970. doi:10.3390/pharmaceutics15030970
307. Filipiak ZM, Bednarska AJ. Different effects of Zn nanoparticles and ions on growth and cellular respiration in the earthworm Eisenia andrei after long-term exposure. Ecotoxicology. 2021;30(3):459–469. doi:10.1007/s10646-021-02360-2
308. Ahmed B, Solanki B, Zaidi A, Khan MS, Musarrat J. Bacterial toxicity of biomimetic green zinc oxide nanoantibiotic: insights into ZnO NP uptake and nanocolloid–bacteria interface. Toxicol Res. 2019;8(2):246–261. doi:10.1039/c8tx00267c
309. Sharma S, Kumar K, Thakur N, Chauhan S, Chauhan MS. The effect of shape and size of ZnO nanoparticles on their antimicrobial and photocatalytic activities: a green approach. Bull Mater Sci. 2020;43:20. doi:10.1007/s12034-019-1986-y
310. Jin SE, Jin JE, Hwang W, Hong SW. Photocatalytic antibacterial application of zinc oxide nanoparticles and self-assembled networks under dual UV irradiation for enhanced disinfection. Int J Nanomed. 2019;14:1737–1751. doi:10.2147/IJN.S192277
311. Abd-Elmaqsoud IG, Elsaadawi HA, Ahmed AI, AbdelKhalek A, Arisha A. The vast biomedical applications of zinc oxide nanoparticles. Zagazig Vet J. 2022;50(3):201–218. doi:10.21608/ZVJZ.2022.144910.1182
312. Alhazmi NM, Sharaf EM. Fungicidal activity of zinc oxide nanoparticles against azole-resistant Aspergillus flavus isolated from yellow and white maize. Molecules. 2023;28(2):711. doi:10.3390/molecules28020711
313. Perveen R, Shujaat S, Qureshi Z, Nawaz S, Khan MI, Iqbal M. Green versus sol-gel synthesis of ZnO nanoparticles and antimicrobial activity evaluation against panel of pathogens. Iran J Microbiol. 2020;9(4):7817–7827. doi:10.1016/j.jmrt.2020.05.004
314. Wahab S, Salman A, Khan Z, Khan S, Krishnaraj C, Yun SI. Metallic nanoparticles: a promising arsenal against antimicrobial resistance-unraveling mechanisms and enhancing medication efficacy. Int J Mol Sci. 2023;24(19):14897. doi:10.3390/ijms241914897
315. Krishnamoorthy R, Athinarayanan J, Periyasamy VS, Alshuniaber MA, Alshammari G, Hakeem MJ, Ahmed MA, Alshatwi AA. Antibacterial mechanisms of zinc oxide nanoparticle against bacterial food pathogens resistant to beta-lactam antibiotics. Molecules. 2022;27(8):2489. doi:10.3390/molecules27082489
316. Huang T, Li X, Maier M, O’Brien-Simpson NM, Heath DE, O’Connor AJ. Using inorganic nanoparticles to fight fungal infections in the antimicrobial resistant era. Acta Biomater. 2023;158:56–79. doi:10.1016/j.actbio.2023.01.019
317. Çetinarslan T, Kümper L, Fölster-Holst R. The immunological and structural epidermal barrier dysfunction and skin microbiome in atopic dermatitis-an update. Front Mol Biosci. 2023;10:1159404. doi:10.3389/fmolb.2023.1159404
318. Kahru A, Mortimer M. Advances in nanotoxicology: towards enhanced environmental and physiological relevance and molecular mechanisms. Nanomaterials. 2021;11(4):919. doi:10.3390/nano11040919
319. Gulab H, Fatima N, Tariq U, Gohar O, Irshad M, Khan MZ, Saleem M, Ghaffar A, Hussain M, Khaliq Jan A, Humayun M, Motola M, Hanif MB. Advancements in zinc oxide nanomaterials: synthesis, properties, and diverse applications. Nano-Struct Nano-Objects. 2024;39:101271. doi:10.1016/j.nanoso.2024.101271
320. Yu H, Lin L, Zhang Z, Zhang H, Hu H. Targeting NF-κB pathway for the therapy of diseases: mechanism and clinical study. Signal Transduct Target Ther. 2020;5(1):209. doi:10.1038/s41392-020-00312-6
321. Liu D, Zhong Z, Karin M. NF-κB: a double-edged sword controlling inflammation. Biomedicines. 2022;10(6):1250. doi:10.3390/biomedicines10061250
322. Wang P, Hu G, Zhao W, Du J, You M, Xv M, Yang H, Zhang M, Yan F, Huang M, Wang X. Continuous ZnO nanoparticle exposure induces melanoma-like skin lesions in epidermal barrier dysfunction model mice through anti-apoptotic effects mediated by the oxidative stress-activated NF-κB pathway. J Nanobiotechnol. 2022;20(1):111. doi:10.1186/s12951-022-01308-w
323. Imraish A, Abu Thiab T, Al‐Awaida W, Al‐Ameer HJ, Bustanji Y, Hammad H, Alsharif M, Al‐Hunaiti A. In vitro anti‐inflammatory and antioxidant activities of ZnFe2O4 and CrFe2O4 nanoparticles synthesized using Boswellia carteri resin. J Food Biochem. 2021;45(6):e13730. doi:10.1111/jfbc.13730
324. Abdelbaky AS, Abd El-Mageed TA, Babalghith AO, Selim S, Mohamed AM. Green synthesis and characterization of ZnO nanoparticles using Pelargonium odoratissimum (L.) aqueous leaf extract and their antioxidant, antibacterial and anti-inflammatory activities. Antioxidants. 2022;11(8):1444. doi:10.3390/antiox11081444
325. Khalaf AA, Hassanen EI, Azouz RA, Zaki AR, Ibrahim MA, Farroh KY, Galal MK. Ameliorative effect of zinc oxide nanoparticles against dermal toxicity induced by lead oxide in rats. Int J Nanomed. 2019;14:7729–7741. doi:10.2147/IJN.S220572
326. Subramaniam VD, Prasad SV, Banerjee A, Gopinath M, Murugesan R, Marotta F, Sun XF, Pathak S. Health hazards of nanoparticles: understanding the toxicity mechanism of nanosized ZnO in cosmetic products. Drug Chem Toxicol. 2019;42(1):84–93. doi:10.1080/01480545.2018.1491987
327. Ingawale DK, Mandlik SK, Patel SS. Anti-inflammatory potential of hecogenin on atopic dermatitis and airway hyper-responsiveness by regulation of pro-inflammatory cytokines. Immunopharmacol Immunotoxicol. 2019;41(2):327–336. doi:10.1080/08923973.2019.1608445
328. Chaturvedi VK, Singh A, Singh VK, Singh MP. Cancer nanotechnology: a new revolution for cancer diagnosis and therapy. Curr Drug Metab. 2019;20(6):416–429. doi:10.2174/1389200219666180918111528
329. Diab T, AlKafaas SS, Shalaby TI, Hessien M. Dexamethasone simulates the anticancer effect of nano-formulated paclitaxel in breast cancer cells. Bioorg Chem. 2020;99:103792. doi:10.1016/j.bioorg.2020.103792
330. Abdelsalam IM, Ghosh S, AlKafaas SS, Bedair H, Malloum A, ElKafas SS, Saad-Allah KM. Nanotechnology as a tool for abiotic stress mitigation in horticultural crops. Biologia. 2023;78(1):163–178. doi:10.1007/s11756-022-01251-z
331. Badıllı U, Mollarasouli F, Bakirhan NK, Ozkan Y, Ozkan SA. Role of quantum dots in pharmaceutical and biomedical analysis, and its application in drug delivery. TRAC Trends Anal Chem. 2020;131:116013. doi:10.1016/j.trac.2020.116013
332. Senapati S, Mahanta AK, Kumar S, Maiti P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct Target Ther. 2018;3(1):7. doi:10.1038/s41392-017-0004-3
333. Prieložná J, Mikušová V, Mikuš P. Advances in the delivery of anticancer drugs by nanoparticles and chitosan-based nanoparticles. Int J Pharm-X. 2024;8:100281. doi:10.1016/j.ijpx.2024.100281
334. Mirón-Barroso S, Domènech EB, Trigueros S. Nanotechnology-based strategies to overcome current barriers in gene delivery. Int J Mol Sci. 2021;22(16):8537. doi:10.3390/ijms22168537
335. Azeem F, Ijaz U, Rashid S, Nadeem H, Manzoor H, Hussain S. Nanomedicine and gene delivery. In: Verpoort F, Ahmad I, Ahmad A, Khan A, Chee CY., editors. Nanomedicine Manufacturing and Applications. Amsterdam, Netherlands: Elsevier; 2021:247–260.
336. Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery systems for nucleic acids and proteins: barriers, cell capture pathways and nanocarriers. Pharmaceutics. 2021;13(3):428. doi:10.3390/pharmaceutics13030428
337. Xiao Y, Shi K, Qu Y, Chu B, Qian Z. Engineering nanoparticles for targeted delivery of nucleic acid therapeutics in tumor. Mol Ther Methods Clin Dev. 2019;12:1–18. doi:10.1016/j.omtm.2018.09.002
338. Cao Y, Tan YF, Wong YS, Liew MWJ, Venkatraman S. Recent advances in chitosan-based carriers for gene delivery. Mar Drugs. 2019;17(6):381. doi:10.3390/md17060381
339. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101–124. doi:10.1038/s41573-020-0090-8
340. Egwu CO, Aloke C, Onwe KT, Umoke CI, Nwafor J, Eyo RA, Chukwu JA, Ufebe GO, Ladokun J, Audu DT, Agwu AO, Obasi DC, Okoro CO. Nanomaterials in drug delivery: strengths and opportunities in medicine. Molecules. 2024;29(11):2584. doi:10.3390/molecules29112584
341. Mondal SK, Chakraborty S, Manna S, Mandal SM. Antimicrobial nanoparticles: current landscape and future challenges. RSC Pharmaceutics. 2024;1(3):388–402. doi:10.1039/D4PM00032C
342. Gadewar M, Prashanth GK, Ravindra Babu M, Dileep MS, Prashanth PA, Rao S, Mahadevaswamy M, Kumar Ghosh M, Singh N, Mandotra SK, Chauhan A, Rustagi S, Yogi R, Chinnam S, Ali B, Ercisli S, Orhan E. Unlocking nature’s potential: green synthesis of ZnO nanoparticles and their multifaceted applications – a concise overview. J Saudi Chem Soc. 2024;28(1):101774. doi:10.1016/j.jscs.2023.101774
343. Taylor E, Webster TJ. Reducing infections through nanotechnology and nanoparticles. Int J Nanomed. 2011;6:1463–1473. doi:10.2147/IJN.S22021
344. Zhang ZY, Xiong HM. Photoluminescent ZnO nanoparticles and their biological applications. Materials. 2015;8(6):3101–3127. doi:10.3390/ma8063101
345. Aravantinou AF, Andreou F, Manariotis ID. Long-term toxicity of ZnO nanoparticles on Scenedesmus rubescens cultivated in semi-batch mode. Nanomaterials. 2020;10(11):2262. doi:10.3390/nano10112262
346. Padmanabhan P, Kumar A, Kumar S, Chaudhary RK, Gulyás B. Nanoparticles in practice for molecular-imaging applications: an overview. Acta Biomater. 2016;41:1–16. doi:10.1016/j.actbio.2016.06.003
347. Farzin L, Sheibani S, Moassesi ME, Shamsipur M. An overview of nanoscale radionuclides and radiolabeled nanomaterials commonly used for nuclear molecular imaging and therapeutic functions. J Biomed Mater Res A. 2019;107(1):251–285. doi:10.1002/jbm.a.36550
348. Zhang Y, Nayak T Hong H Cai W. Biomedical applications of zinc oxide nanomaterials. Curr Mol Med. 2013;13(10):1633–1645. doi:10.2174/1566524013666131111130058
349. Lei G, Yang S, Cao R, Zhou P, Peng H, Peng R, Zhang X, Yang Y, Li Y, Wang M, He Y. In situ preparation of amphibious ZnO quantum dots with blue fluorescence based on hyperbranched polymers and their application in bio-imaging. Polymers. 2020;12(1):144. doi:10.3390/polym12010144
350. Masar M, Ali H, Guler AC, Urbanek M, Urbanek P, Hanulikova B, Pistekova H, Annusova A, Machovsky M, Kuritka I. Multifunctional bandgap-reduced ZnO nanocrystals for photocatalysis, self-cleaning, and antibacterial glass surfaces. Colloids Surf A Physicochem Eng Asp. 2023;656:130447. doi:10.1016/j.colsurfa.2022.130447
351. Xie J, Li H, Zhang T, Song B, Wang X, Gu Z. Recent advances in ZnO nanomaterial-mediated biological applications and action mechanisms. Nanomaterials. 2023;13(9):1500. doi:10.3390/nano13091500
352. Ali AA, Altemimi AB, Alhelfi N, Ibrahim SA. Application of biosensors for detection of pathogenic food bacteria: a review. Biosensors. 2020;10(6):58. doi:10.3390/bios10060058
353. Sruthi S, Millot N, Mohanan PV. Zinc oxide nanoparticles mediated cytotoxicity, mitochondrial membrane potential and level of antioxidants in presence of melatonin. Int J Biol Macromol. 2017;103:808–818. doi:10.1016/j.ijbiomac.2017.05.088
354. Cho IH, Kim DH, Park S. Electrochemical biosensors: perspective on functional nanomaterials for on-site analysis. Biomater Res. 2020;24:6. doi:10.1186/s40824-019-0181-y
355. Upadhyay PK, Jain VK, Sharma K, Sharma R. Synthesis and applications of ZnO nanoparticles in biomedicine. Res J Pharm Technol. 2020;13(4):1636–1644. doi:10.5958/0974-360X.2020.00297.8
356. Dönmez S. Green synthesis of zinc oxide nanoparticles using Zingiber officinale root extract and their applications in glucose biosensor. El-Cezeri. 2020;7(3):1191–1200. doi:10.31202/ecjse.729462
357. Krishna MS, Singh S, Batool M, Fahmy HM, Seku K, Shalan AE, Lanceros-Mendez S, Zafar MN. A review on 2D-ZnO nanostructure-based biosensors: from materials to devices. Mater Adv. 2023;4(2):320–354. doi:10.1039/D2MA00878E
358. Shetti NP, Bukkitgar SD, Reddy KR, Reddy CV, Aminabhavi TM. ZnO-based nanostructured electrodes for electrochemical sensors and biosensors in biomedical applications. Biosens Bioelectron. 2019;141:111417. doi:10.1016/j.bios.2019.111417
359. Zhang N, Xiong G, Liu Z. Toxicity of metal-based nanoparticles: challenges in the nano era. Front Bioeng Biotechnol. 2022;10:1001572. doi:10.3389/fbioe.2022.1001572
360. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49(11):1603–1616. doi:10.1016/j.freeradbiomed.2010.09.006
361. Saptarshi SR, Duschl A, Lopata AL. Biological reactivity of zinc oxide nanoparticles with mammalian test systems: an overview. Nanomedicine. 2015;10(13):2075–2092. doi:10.2217/nnm.15.44
362. Soni D, Gandhi D, Tarale P, Bafana A, Pandey RA, Sivanesan S. Oxidative stress and genotoxicity of zinc oxide nanoparticles to Pseudomonas species, human promyelocytic leukemic (HL-60), and blood cells. Biol Trace Elem Res. 2017;178:218–227. doi:10.1007/s12011-016-0921-y
363. Kaushal P, Maity D, Awasthi R. Nano-green: harnessing the potential of plant extracts for sustainable antimicrobial metallic nanoparticles. J Drug Deliv Sci Technol. 2024;94:105488. doi:10.1016/j.jddst.2024.105488
364. Rasmiya begum SL, Jayawardana NU. Green synthesized metal nanoparticles as an ecofriendly measure for plant growth stimulation and disease resistance. Plant Nano Biol. 2023;3:100028. doi:10.1016/j.plana.2023.100028
365. Maocong H, Zhenhua Y, Xianqin W. Characterization techniques for graphene-based materials in catalysis. AIMS Mater Sci. 2017;4(3):755–788. doi:10.3934/matersci.2017.3.755
366. Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology’s frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther. 2024;9(1):34. doi:10.1038/s41392-024-01745-z
367. Narayanan M, Devi D, Kandhasamy S, Gnanasekaran C, Govindhan R, Manoharan N. Role of bioactive compounds synthesized by extremophilic microbes and their bioactivity. In: Mérillon JM, Ramawat KG., editors. Plant Specialized Metabolites: Phytochemistry, Ecology and Biotechnology. Cham, Switzerland: Springer; 2024:1–24. doi:10.1007/978-3-031-30037-0_54-1
© 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.