Next Article in Journal
MRE Encapsulating MRG: Synergistic Improvement in Modulus Tunability and Energy Dissipation
Previous Article in Journal
Effect of Ga2O3 Content on the Activity of Al2O3-Supported Catalysts for the CO2-Assisted Oxidative Dehydrogenation of Propane
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Zinc Oxide Nanoparticles as Next-Generation Feed Additives: Bridging Antimicrobial Efficacy, Growth Promotion, and Sustainable Strategies in Animal Nutrition

Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China
*
Author to whom correspondence should be addressed.
Nanomaterials 2025, 15(13), 1030; https://doi.org/10.3390/nano15131030
Submission received: 27 May 2025 / Revised: 16 June 2025 / Accepted: 30 June 2025 / Published: 2 July 2025
(This article belongs to the Section Environmental Nanoscience and Nanotechnology)

Abstract

Zinc oxide nanoparticles (ZnO NPs) have attracted significant attention due to their wide-ranging applications in animal production, largely because of their notable biocompatibility, low toxicity, and strong antimicrobial activity. These properties make ZnO NPs a promising substitute for traditional antibiotics. Their application could address the growing concern of antibiotic resistance in livestock industries. This review examines the unique physicochemical characteristics of ZnO NPs, including their nanoscale size and high surface area, which contribute to their biological functionality. Emphasis is placed on green synthesis methods that minimize environmental impact while producing high-quality ZnO NPs. In animal farming, ZnO NPs play a crucial role not only in promoting growth and improving immune responses, but also in enhancing meat and egg quality. Additionally, this review discusses the environmental and safety implications of ZnO NPs, considering their sustainable application potential in future animal production practices, aimed at fostering a more eco-friendly and efficient livestock sector.

Graphical Abstract

1. Introduction

As the antibiotic resistance crisis intensifies, the global livestock industry faces unprecedented challenges. The overuse of antibiotics has not only led to the spread of resistant bacteria but also poses significant threats to animal health and human food safety. Therefore, finding alternatives to antibiotics has become an urgent issue in animal production. The rapid development of nanotechnology has provided new solutions to this problem, with nano-sized zinc oxide (ZnO NPs) showing unique potential in the field of animal nutrition.
Due to its excellent biocompatibility, high surface activity, and multifunctionality, ZnO NPs have become a hot topic in recent research. The small size of ZnO NPs (ranging from 1 to 100 nm) gives them a large specific surface area, which allows for more efficient absorption and utilization in animals [1,2,3]. In addition, ZnO NPs not only exhibit strong antibacterial activity but also effectively regulate the immune system, promote growth, and enhance animals’ tolerance to oxidative stress [4,5]. These characteristics make ZnO NPs an ideal candidate for replacing antibiotics, particularly in reducing antibiotic use, improving animal health, and enhancing production performance [6,7].
While this review emphasizes biosynthesized ZnO NPs for animal nutrition, their synthesis method critically impacts applicability. Biosynthesis offers environmental advantages through green chemistry and may impart bioactive surface modifications. However, it typically yields NPs with lower production, reduced stability, and colloidal handling requirements, contrasting with conventional methods’ higher yields and particle control [8]. Despite these challenges, biosynthesized NPs’ unique properties drive optimization efforts. Key differences in properties and functionality will be detailed in Section 2.1 and Section 2.2.
Beyond their advantages in improving animal performance and health, ZnO NPs also show potential for environmental sustainability. Compared to traditional zinc supplements, ZnO NPs have higher bioavailability, which means that they can meet animals’ zinc requirements at lower doses, thereby reducing excessive zinc discharge into the environment and mitigating ecological risks [9].
However, despite the promising prospects of ZnO NPs in animal nutrition, there are still gaps in the current research. First, the optimization of ZnO NP dosage remains to be addressed, ensuring that their use improves animal performance while avoiding the negative effects of overuse. Secondly, there is a lack of sufficient studies on the long-term safety of ZnO NPs, particularly regarding the potential risks to animal health with prolonged exposure. Furthermore, the behavior, ecological toxicity, and mechanisms of ZnO NPs in the environment have yet to be fully explored, which are key factors determining their feasibility and sustainability in practical applications.
Therefore, this paper aims to explore the potential of ZnO NPs in animal nutrition, with a focus on their antibacterial, immune-regulatory, and growth-promoting effects. Additionally, it highlights the current research gaps and challenges, providing a reference for future studies in this area.

2. Green Synthesis and Physicochemical Tailoring of ZnO NPs

2.1. Green Synthesis Methods (Plant-/Microbe-Mediated) and Their Environmental Benefits

Compared with traditional chemical synthesis methods, green synthesis not only reduces the use of harmful chemical reagents, but also significantly reduces the environmental impact, which has obvious advantages in terms of environmental protection and sustainable development. The green synthesis method for ZnO nanoparticles, using plant or microbial extracts (such as bacteria and fungi) as stabilizers to effectively reduce environmental toxicity, is shown in Figure 1. These natural extracts are rich in phenols, alcohols, and terpene compounds, which help to stabilize the nanoparticles and promote the synthesis of high-quality, stable ZnO nanoparticles [10,11]. Plant extracts, bacteria, and fungi can efficiently synthesize ZnO nanoparticles after reacting with zinc precursors and bioactive molecules. However, it is important to note that the chemical properties of ZnO NPs obtained via these green methods can vary significantly depending on the biological source used. For example, plant extracts and microorganisms (bacteria and fungi) provide diverse organic molecules that ‘decorate’ the surface of the nanoparticles, which in turn affects their chemical properties and, likely, their biological activities. This chemical diversity should be carefully considered, as it may influence the NPs’ antimicrobial efficacy, immune modulation, and other biological functions. Complete chemical characterization of the synthesized nanoparticles (NPs) is essential to understand their composition and behavior. Techniques like spectroscopy, chromatography, and surface charge analysis help identify the functional groups and chemical signatures on the nanoparticle surfaces. Standardizing the synthesis process is also important to ensure consistency between batches, ensuring reproducibility and reliable results in both research and practical applications such as animal feed additives or therapeutic agents.

2.2. Comparison of ZnO Nanoparticles with Conventional Zinc Oxide: Bioavailability, Ecotoxicity, and Environmental Impact

Zinc oxide nanoparticles (ZnO NPs) possess distinctive physicochemical properties, particularly their nanoscale dimensions (1–100 nm) and exceptionally high surface-to-volume ratio, which collectively contribute to their superior bioavailability when compared to conventional zinc oxide and organic zinc compounds [12]. The unique nanoscale characteristics of ZnO NPs facilitate enhanced permeability across biological barriers, enabling efficient penetration into cellular and tissue structures. This exceptional property renders them highly advantageous for diverse applications, including targeted drug delivery systems, antimicrobial formulations, and innovative agricultural solutions. In contrast, conventional zinc oxide, despite its effectiveness in specific applications, often necessitates higher concentrations to achieve comparable efficacy. Moreover, the synthesis processes of traditional zinc oxide may yield toxic by-products, posing significant risks of environmental contamination and adverse effects on non-target organisms [13]. The controlled release mechanism of Zn2+ ions from ZnO NPs represents a significant advancement in environmental safety, as it effectively regulates zinc ion availability, thereby minimizing the risk of excessive zinc accumulation in ecosystems and substantially reducing potential ecological toxicity [13]. Furthermore, recent developments in green synthesis methodologies utilizing plant extracts or microbial systems have demonstrated promising results in producing ZnO NPs with reduced ecotoxicity. These environmentally benign synthesis approaches not only mitigate pollution but also enhance the biodegradability of nanoparticles, resulting in significantly lower ecological impact compared to conventional zinc sources [10,11].

3. ZnO NPs as Multifunctional Feed Additives: Mechanisms and Applications

3.1. Antibacterial Action

Studies have shown that ZnO NPs inhibit the growth of Escherichia coli and Klebsiella pneumoniae at a concentration of 30 μg/mL, which is the highest antibacterial activity [14]. Although the antimicrobial activity of ZnO nanoparticles (NPs) is widely recognized, the exact mechanisms underlying their toxicity remain not fully elucidated and continue to be debated. ZnO NPs exhibit broad-spectrum antibacterial activity against both Gram-positive and Gram-negative bacteria, with a notably stronger inhibitory effect on Gram-negative strains, extending even to heat-resistant and pressure-tolerant spores [15,16]. Nanoparticles with antimicrobial properties can penetrate the peptidoglycan layer and cause significant damage to bacterial cells. The presence of the lipopolysaccharide layer in the bacterial cell wall enhances direct contact between the bacterial membrane and nanoparticles. This interaction occurs because lipopolysaccharides carry a negative charge, attracting and promoting the absorption of metal ions from positively charged nanoparticles, ultimately leading to bacterial cell damage [17]. Recent research indicates significant differences in sensitivity based on bacterial type: Gram-negative pathogens such as E. coli and Pseudomonas aeruginosa exhibit 100% growth inhibition (IC100) at 0.6 mM ZnO NPs, whereas Gram-positive strains, including Staphylococcus aureus and Bacillus subtilis, require higher concentrations (~1.0 mM) for equivalent inhibition [18]. Critically, exposure to these inhibitory concentrations for as little as 15 min induces substantial membrane damage in over 70% of bacterial cells, demonstrating rapid bactericidal action through membrane targeting. Several mechanisms have been proposed, including physical interactions between ZnO nanoparticles and the bacterial cell wall [19], the generation of reactive oxygen species (ROS) [20,21], and the release of free radicals and Zn2+ ions [22]. As illustrated in (Figure 2), the general mechanisms of ZnO nanoparticle antimicrobial action can be summarized as follows:
(1) Nanoparticles aggregate on the cell surface, causing membrane rupture and leakage of cellular contents [23,24].
(2) Bacterial cells absorb the released Zn2+ ions, leading to a significant decrease in cellular energy levels and DNA damage due to the loss of energy-rich molecules [25].
(3) ROS generation damages cellular components, thereby impairing essential cellular functions [26].
Among all inorganic photocatalytic materials, ZnO stands out due to its high photocatalytic efficiency, particularly its ability to strongly absorb ultraviolet (UV) light [27]. ZnO exhibits excellent responsiveness to UV light, leading to a significant enhancement in electrical conductivity, which notably facilitates its interaction with bacteria. In aqueous solutions, ZnO nanoparticles (NPs) exhibit phototoxicity under UV exposure, generating reactive oxygen species (ROS) such as hydrogen peroxide (H2O2) and superoxide anions (O2−) [28]. Ultimately, the bacterial cell membrane barrier is compromised by H2O2, leading to the disruption of cellular components such as DNA, proteins, and lipids, which results in cell damage and death. This process has driven the use of ZnO NPs in numerous antibacterial applications within the fields of biotechnologies and biomedical nanotechnology.
The antimicrobial efficacy of ZnO nanoparticles is influenced by various factors, including size, shape, zeta potential, surface morphology, surface defects, charge, functionalization, and other physicochemical properties. Additionally, environmental factors, microbial type, exposure time, and growth medium significantly affect the antibacterial activity of ZnO nanoparticles [29]. Some studies suggest that increased surface area-to-volume ratio, ligand deficiency, and enhanced surface energy contribute to the accumulation of metal oxide nanoparticles on the cell surface [30]. These studies have shown that the antibacterial properties of nanoparticles can be significantly improved by changing their surface properties, which provides new ideas for the development of more effective antibacterial agents. In addition, this discovery has also promoted the application of nanotechnology in agriculture, food safety, and medical fields, especially in terms of disease prevention as a feed additive, which is of great practical significance.

3.2. Anti-Inflammatory and Antioxidant Regulation

3.2.1. Anti-Inflammatory Effects: Reduction of Inflammatory Reactions and Tissue Damage

ZnO NPs have demonstrated significant anti-inflammatory potential through multiple molecular pathways, primarily by modulating key inflammatory mediators and signaling cascades. An overview of the different anti-inflammatory mechanisms adopted by ZnO NPs is shown (Figure 3). The nanoscale characteristics of ZnO NPs, particularly their high surface area-to-volume ratio, enhance their interaction with cellular components compared to bulk materials, leading to more effective suppression of inflammatory responses [31]. In addition, the interaction of zinc oxide nanoparticles (ZnO NPs) with serum proteins forms protein crowns, which regulate their interaction with cells and immune responses, thereby affecting their anti-inflammatory effects [32].
The anti-inflammatory activity of ZnO nanoparticles is regulated by three main mechanisms: (1) regulation of cytokine production and the NF-κB signaling pathway, (2) inhibition of the degranulation of mast cells, and (3) modulation of inflammatory enzyme activity. In the NF-κB pathway, ZnO NPs effectively inhibit LPS-induced activation by upregulating A20 expression and preventing nuclear translocation of NF-κB p65 in macrophages, while maintaining cytoplasmic levels of IκBα [33].
Regarding mast cell regulation, ZnO NPs demonstrate inhibitory effects by reducing thymic stromal lymphopoietin (TSLP) production and modulating p53 protein levels, thereby controlling the release of pro-inflammatory mediators such as IL-13 and HMGB1 [31]. Furthermore, ZnO NPs exhibit dose-dependent inhibition of key inflammatory enzymes, including COX-2 and iNOS. This enzymatic regulation prevents the production of inflammatory prostaglandins (PGE2) and excessive nitric oxide, which are associated with tissue damage and inflammation amplification [34,35,36].
ZnO NPs also regulate inflammation by reducing the production of NO. NO is a gas molecule produced by nitric oxide synthase (NOS) and plays an important role in many inflammatory diseases. ZnO NPs have been shown to effectively inhibit the expression of NOS and reduce the production of NO, thereby reducing the inflammatory response caused by NO [36]. In addition, ZnO NPs inhibit the catalysis of H2O2 to hypochlorous acid by inhibiting the activity of myeloperoxidase (MPO), thereby reducing the production of ROS and nitrating agents [32].
The multifaceted anti-inflammatory properties of ZnO NPs, encompassing cytokine regulation, cellular proliferation control, and enzymatic activity modulation, position them as promising candidates for therapeutic applications in inflammation-related disorders. Their ability to simultaneously target multiple inflammatory pathways offers potential advantages over conventional anti-inflammatory agents.

3.2.2. Antioxidant Properties: Reducing Oxidative Stress and Cell Damage

Zinc oxide nanoparticles (ZnO NPs) demonstrate significant antioxidant capabilities through multiple mechanisms, both in extracellular environments and intracellular systems. While direct intracellular reactive oxygen species (ROS) scavenging remains unconfirmed, ZnO NPs exhibit antioxidant activity primarily through the modulation of antioxidant enzyme systems [37].
In vitro studies utilizing DPPH assays have established the free radical scavenging capacity of ZnO NPs, with electron density transfer from oxygen to nitrogen atoms resulting in decreased absorbance at 517 nm [38]. This antioxidant activity appears to be concentration-dependent and may be enhanced by surface-adsorbed phytochemicals [39]. Comprehensive evaluations using standardized assays (TAC, TRP, DPPH, and ABTS) have consistently demonstrated concentration-dependent antioxidant activity, with particularly strong performance in ABTS assays [40,41,42,43,44,45].
In vivo investigations have further substantiated these findings. Dietary supplementation with ZnO NPs (39.2–49.11 nm) at concentrations ranging from 5 to 80 ppm has shown significant enhancement of antioxidant enzyme activities (SOD, CAT, and GPx) and a reduction in oxidative stress markers (MDA and lipid peroxides) in various animal models [46,47,48]. Notably, these studies have observed increased mRNA expression of key antioxidant enzymes (SOD1, CAT, GPX1, and GPX7) in multiple tissue types.
The antioxidant properties of ZnO NPs, combined with their antimicrobial and anti-inflammatory activities, contribute to their therapeutic potential in wound healing, immune modulation, and cellular protection. Table 1 provides a comprehensive overview of the mechanisms by which ZnO NPs exert these effects, highlighting their multifunctional roles in pathogen elimination, inflammation reduction, and oxidative stress mitigation.

3.3. Growth Performance and Product Quality Improvements

In the domain of animal production, zinc oxide nanoparticles (ZnO NPs) have emerged as a significant additive, owing to their distinctive physicochemical characteristics. These particles have been the focus of extensive research and application due to their antimicrobial, antioxidant, immunomodulatory, and growth-promoting properties. A substantial body of research has demonstrated that the judicious administration of ZnO NPs has the capacity to enhance immune function in animals, thereby promoting enhanced productivity and the quality of meat, eggs, milk, and other products. This is achieved by means of improving intestinal health and promoting nutrient absorption.
The specific application effects and research progress of ZnO NPs in different animal species are listed in Table 2, covering a wide range of animal models, such as cattle, sheep, pigs, and poultry. From the experimental results, the application of ZnO NPs showed significant positive effects in different animals, especially in disease resistance, immune enhancement, and growth promotion. Furthermore, the bioavailability and environmental impact of ZnO NPs have garnered mounting interest, with future research potentially focusing on optimizing their dosage, enhancing their environmental sustainability, and reducing potential toxic effects.

3.3.1. Therapeutic Effect

Zinc oxide nanoparticles (ZnO NPs) exhibit superior therapeutic efficacy compared to conventional zinc sources, which is attributed to their nanoscale dimensions, high surface area, and enhanced bioavailability [58,59]. These properties enable effective antimicrobial action at lower concentrations while maintaining environmental compatibility.
The antimicrobial mechanism of ZnO NPs involves membrane disruption, leading to cellular content leakage and bacterial death [60]. Their efficacy against both Gram-positive and Gram-negative bacteria is well-documented, with size-dependent activity observed against Staphylococcus aureus and concentration-dependent inhibition of Escherichia coli O157 [16,61].
In veterinary applications, ZnO NPs demonstrate broad-spectrum activity against gastrointestinal pathogens. Notably, they exhibit dose-dependent antifungal effects against Candida albicans, including fluconazole-resistant strains, at concentrations of 119 μg/mL [62,63,64]. For reproductive health, biosynthesized ZnO NPs have shown remarkable efficacy in treating postpartum endometritis in dairy cows, significantly reducing uterine pathogens and improving reproductive outcomes [52].
In poultry production, ZnO NPs effectively combat respiratory pathogens when combined with tylosin, while reducing antibiotic residues [65]. Their antiparasitic properties have been demonstrated against Eimeria flexneri in broilers and Toxoplasma gondii in murine models, showing reduced pathogen load and improved tissue integrity [66,67].
These findings position ZnO NPs as promising therapeutic agents for various veterinary applications, offering advantages in efficacy, reduced dosage requirements, and minimized resistance development.

3.3.2. Immunomodulatory Effects of Zinc Oxide Nanoparticles in Animal Production

Terrestrial Livestock Applications
Zinc oxide nanoparticles (ZnO NPs) serve as potent immunomodulators in terrestrial livestock, enhancing both innate and adaptive immunity through multiple mechanisms. Their nanoscale properties (high surface area and bioavailability) improve zinc utilization efficiency compared to conventional sources, addressing zinc deficiency-related immune dysfunction [68,69]. In swine production, dietary supplementation with 800 mg/kg ZnO NPs increases average daily weight gain by 18%, reduces diarrhea incidence by 40%, and enhances antioxidant capacity through elevated serum SOD (↑32%) and GSH-Px (↑25%) activities [49,70]. The nanoparticles strengthen intestinal barrier integrity via upregulation of tight junction proteins (ZO-1 and occludin) and modulate gut microbiota composition, suppressing pathogenic Escherichia coli K88 while promoting beneficial microbial communities [71,72].
Poultry studies demonstrate ZnO NPs’ dual role in immune organ development and pathogen control. Supplementation increases thymus (↑15%), bursa (↑12%), and spleen (↑9%) weights, correlating with enhanced antimicrobial activity against Salmonella enterica and Escherichia coli O157 [73,74,75,76]. The size-dependent antibacterial efficacy (40–60 nm particles showing optimal performance) arises from membrane disruption and reactive oxygen species generation [19,77]. Furthermore, ZnO NPs regulate stress-responsive genes, modulating HSP70/90 expression in intestinal tissues and appetite-related hormones (CCK and gastrin), thereby improving stress resilience [78].
Aquatic Species Applications
In aquaculture systems, ZnO NPs exhibit superior immunostimulatory effects over bulk zinc, achieving equivalent biological responses at 60% lower dosages (40–60 mg/kg feed) [79,80,81]. Evidence suggests that ZnO NPs enhance the immune response in aquatic animals by modulating cytokine gene expression and promoting non-specific receptor-mediated cell development, including neutrophils. This creates a strong first line of defense against infections and foreign pathogens, such as bacteria [79,82,83]. Furthermore, ZnO NPs have demonstrated potent antimicrobial activity at low doses. Studies have shown that the addition of 40–60 mg/kg of ZnO NPs to fish feed significantly improves immune performance and disease resistance, effectively preventing infections [51,84,85]. Their eco-friendly profile minimizes zinc accumulation in aquatic ecosystems while maintaining therapeutic efficacy. This immunomodulatory capacity, coupled with growth-promoting and antimicrobial properties, positions ZnO NPs as multifunctional additives for sustainable animal production systems.

3.3.3. Growth Promotion and Productivity Improvement

In recent years, the incorporation of nanotechnology, particularly zinc oxide nanoparticles (ZnO NPs), into animal feed has attracted considerable attention. These nanoparticles have been shown to enhance the bioavailability of trace minerals, with notable benefits observed in weaned piglets and poultry. ZnO NPs not only support improved growth and development but also enhance feed conversion efficiency, leading to significant economic advantages [86,87].
Growth Promotion Mechanisms and Feed Efficiency Optimization
A study by Qu demonstrated that supplementing piglet diets with small-sized zinc oxide nanoparticles (ZnO NPs, average size: 21 nm) significantly improved daily weight gain, the feed conversion ratio (FCR), and the lymphoid organ index. Additionally, ZnO NPs enhanced intestinal health, as evidenced by increased villus height and a higher villus height-to-crypt depth (VH/CD) ratio [77]. Furthermore, ZnO NPs have been shown to reduce somatic cell counts and increase milk production in cows suffering from subclinical mastitis, indicating their potential as both a preventive and therapeutic agent for managing this condition in dairy cows [88].
In vitro studies have also highlighted the effects of ZnO NPs on rumen microorganisms. The addition of ZnO NPs (100–200 mg/kg) to rumen contents during the early stages of incubation (6 to 12 h) promoted microbial growth, enhanced microbial protein synthesis, and improved energy utilization efficiency. Specifically, these nanoparticles increased concentrations of volatile fatty acids, microbial crude protein production, and organic matter fermentation. However, they also led to higher ammonia–nitrogen concentrations and altered the acetic acid-to-propionic acid ratio [89].
Improving Poultry Performance
The use of zinc oxide nanoparticles (ZnO NPs) in poultry production has shown considerable benefits. Broiler diets often contain high levels of phytates, which can impair zinc absorption [90]. In comparison with both organic and inorganic zinc sources, the addition of 80 mg/kg of ZnO NPs has proven to be the most effective in improving broiler performance and immune function [91]. A study by Abeer demonstrated that supplementing broiler diets with 90 mg/kg of ZnO NPs significantly increased body weight and sustained weight gain throughout the trial, outperforming conventional bulk zinc oxide [92].
Furthermore, the inclusion of 20–60 mg/kg of ZnO NPs in laying hen diets enhanced nutrient digestibility and improved liver and kidney function [93]. In broiler chickens, supplementation with 30–90 mg/kg of ZnO NPs led to notable increases in weight gain, feed intake, and overall growth performance [73]. Ahmed’s research found that adding 20, 40, and 60 mg/kg of ZnO NPs to broiler rations under hot environmental conditions improved growth performance, nutrient digestibility, carcass quality, and liver and kidney function [94]. Similarly, Abbasi reported that ZnO NP supplementation significantly boosted quail fertility, with peak egg production achieved at dietary zinc levels of 67–72 mg/kg, whether using ZnO NPs or zinc oxide microparticles [53].
Effects on Meat and Egg Quality
Zinc oxide nanoparticles (ZnO NPs) demonstrate enhanced bioavailability and reduced dosage requirements compared to conventional zinc supplements. Studies demonstrate that ZnO NPs exert beneficial effects on multiple poultry performance parameters, including carbonic anhydrase activity optimization, feed conversion efficiency enhancement, and improvements in egg quality indices [95,96,97]. Significant increases in serum zinc levels, eggshell zinc deposition, and tibial bone mineralization have been documented [98], along with enhanced antioxidant capacity in biological systems [96,99]. Dietary supplementation with ZnO NPs has been demonstrated to enhance eggshell strength, intensify yolk coloration, and elevate egg zinc content, thereby improving nutritional value [100].
Safdar conducted a comparative analysis of ZnO NPs synthesized through various green chemistry approaches, evaluating their effects on laying hen productivity, skeletal integrity, and oxidative stress responses. While nanoparticle synthesis methods showed no significant performance differences, optimal production parameters were achieved at a 70 mg/kg ZnO NP concentration [101]. Complementary research by Usama revealed that broiler diets supplemented with 20 mg/kg ZnO NPs significantly improved carcass characteristics, including enhanced dressing percentage, edible meat yield, and total carcass weight [102,103,104,105].
In porcine models, ZnO NP supplementation improved meat quality through enhanced water retention capacity. These improvements were mechanistically linked to Nrf2-GCL pathway activation, which mitigates oxidative damage while improving myofibrillar structure and tissue integrity [106].
Despite these advantages, emerging evidence indicates that high concentrations of nanometer-scale zinc oxide may raise toxicological concerns. Cytotoxicity assessments in avian models have demonstrated a dose-dependent relationship between exposure and hematological abnormalities, including erythrocyte nuclear mutations and binucleated cell formation [107]. Contrastingly, Yusuf reported decreased bone mineralization indices and compromised eggshell quality in nano-ZnO groups, suggesting potential interference with calcium–phosphate metabolism [108]. These findings underscore the critical balance between ZnO NPs’ zootechnical benefits and their toxicological profile, emphasizing the necessity for precise dosage optimization to prevent counterproductive effects on poultry growth performance and product quality.

3.4. Reproductive Health and Genetic Enhancement

A major challenge in livestock production is the limited availability of high-quality breeding stock, where the presence of genetically superior reproductive males can significantly enhance overall herd productivity. Zinc plays a crucial role in sperm motility by regulating the ATP system within phospholipid energy reserves, while its deficiency increases oxidative stress, leading to diminished sperm quality. Moreover, oxidative damage caused by reactive oxygen species during sperm cryopreservation can impair motility, an effect that can be effectively mitigated using antioxidant nanoparticles such as zinc oxide nanoparticles (ZnO NPs). Abedin demonstrated that ZnO NPs more effectively prevented oxidative damage to stag spermatozoa compared to selenium nanoparticles (Se-NPs) and improved semen quality by upregulating HSP gene expression [109]. Additionally, ZnO NPs synthesized by Leila from mulberry–orange fruit extracts significantly enhanced the survival, viability, and DNA integrity of ram spermatozoa during cryopreservation at 4 °C [110]. Motlagh further reported that supplementing sperm storage media with 100 μg/mL of zinc nanoparticles and ZnO NPs successfully preserved the quality of rooster semen during the cooling period [111].

4. Dissolution and Bioavailability of Zinc Oxide Nanoparticles in Acidic Environments

When adding zinc oxide nanoparticles (ZnO NPs) to animal feed, controlling dissolution kinetics presents a challenging issue. ZnO NPs exhibit unique dissolution properties in acidic environments, such as the gastrointestinal (GI) tract, where the pH typically ranges from 1 to 4, significantly affecting their bioavailability. This dissolution is influenced by various factors, including the particle size, specific surface area, and aggregation state of the zinc oxide nanoparticles [112]. Smaller particle sizes and larger surface areas typically increase the dissolution rate, as they provide more surface area for interaction with the acidic medium. However, altering the feed matrix can affect the dissolution of ZnO in the gastrointestinal tract. The presence of various biological matrices such as proteins, carbohydrates, and lipids may influence the dissolution process. Depending on their chemical nature and interactions with the nanoparticles, these matrices can either promote or hinder the release of zinc ions. For example, circular dichroism studies found that the conformation of ZnO NPs changed after interaction with skim milk and casein. After 1 min of exposure to ZnO NPs, slight deformation of the helical bands was observed, and after 168 h of exposure, no significant deformation was observed, indicating that proteins can bind to zinc oxide nanoparticles and alter their surface properties, thus influencing the dissolution rate and subsequent bioavailability [113]. Furthermore, bioavailability is not solely dependent on the dissolution rate. Other factors, including the presence of competing minerals such as calcium or phytates, can also affect the efficiency of zinc absorption [114]. Therefore, understanding how these factors interact to influence the dissolution and absorption processes of zinc oxide nanoparticles is crucial for optimizing their use in animal feed.

5. Toxicity and Safety Considerations

Although the addition of metal nanoparticles to animal diets has potential applications, the long-term effects of their use as feed additives remain largely unknown. At present, the toxicological data on nanoparticles are incomplete and their potential hazards need to be thoroughly assessed. Therefore, before nanoparticles can be officially recommended for use in animal nutrition, extensive long-term animal studies must be conducted to evaluate their toxicity characteristics and establish a safe range of use to ensure their safety and reliability in the livestock industry. Recent advances in omics approaches, such as transcriptomics, proteomics, and metabolomics, offer powerful tools to unravel the molecular mechanisms of nanoparticle toxicity. These methods provide comprehensive insights into gene expression changes, protein dysregulation, and metabolic pathway alterations, enabling a deeper understanding of nanomaterial interactions with biological systems beyond traditional cytotoxicity assays [115]. For instance, transcriptomic studies (e.g., RNA-Seq) in hepatic cells have shown that ZnO NPs significantly upregulate and downregulate genes involved in immune responses and oxidative stress, while metabolomic analyses in zebrafish models reveal disruptions in the tricarboxylic acid cycle and oxidative phosphorylation, leading to ATP loss and energetic stress. These omics techniques have identified key biomarkers, such as reactive oxygen species (ROS) and inflammatory cytokines, which mediate ZnO NP-induced damage and highlight dose-dependent effects [115].
Abdelnour’s study found that 40 mg/kg ZnO NPs significantly increased creatinine levels and the activity of AST, ALT, and CK-MB, suggesting that 40 mg/kg ZnO NPs may have toxic effects on multiple organ systems, including the kidneys, liver, and heart, leading to organ dysfunction [116]. Other research has shown that ZnO NPs, through the release of Zn2+ ions, significantly reduced the survival rate of A172 cells within 24 h, induced DNA damage, and significantly inhibited the movement of zebrafish embryos, suggesting that Zn2+ ions play a key role in their cytotoxicity, genotoxicity, and behavioral toxicity [117]. These findings highlight the need for caution in the use of nanoparticles in animal feed and emphasize the need for further safety evaluations. The integration of omics data, particularly proteomics and lipidomics, has further elucidated that ZnO NPs disrupt protein homeostasis and lipid metabolism in macrophages, contributing to inflammatory responses and cellular damage [115]. Such mechanistic insights are crucial for developing safer nanoparticle formulations and refining dosage regimens to mitigate environmental and health risks.
The mechanism of toxicity of ZnO nanoparticles is shown in Figure 4 and mainly involves oxidative stress and the production of reactive oxygen species leading to cell damage [118]. ZnO NPs dissolve in acidic environments, releasing Zn2+ ions which, together with their semiconducting properties, increase intracellular oxidative stress [118]. This in turn causes DNA and mitochondrial damage, ultimately leading to cell apoptosis or necrosis [118]. In addition, ZnO NPs can activate inflammatory responses, increasing the expression of inflammatory factors such as IL-1β, TNF-α, and IFN-γ, which exacerbate tissue damage [118]. They also alter gene expression related to metabolism, repair, and immune responses, including upregulation of the CYP450 family of enzymes, further enhancing cytotoxic effects [118]. ZnO NPs may also induce a novel mechanism of vascular endothelial cell damage through ferritin autophagy and ferroptosis, in which the AMPK-ULK1 pathway plays a critical role [119]. In particular, NCOA4-mediated ferritin autophagy degradation is a key step in ZnO NP-induced ferroptosis [119].

6. Environmental Fate and Ecotoxicity of Zinc Oxide Nanoparticles in Animal Feed

As the use of ZnO NPs in animal feed continues to increase, it is crucial to assess their long-term ecological risks. Once incorporated into animal diets, ZnO NPs are typically excreted, with a significant portion entering the environment, posing potential threats to aquatic and soil ecosystems. Upon release into the environment, ZnO NPs undergo various transformations, such as dissolution, aggregation, or sedimentation. These processes are influenced by factors such as pH, ionic strength, and dissolved organic matter [120]. Smaller ZnO NPs typically exhibit higher bioavailability and greater toxicity to aquatic organisms. Moreover, aggregation and coagulation in aquatic environments can alter the size distribution of ZnO NPs, subsequently affecting their toxicity [121].
The ecotoxicity of ZnO NPs in aquatic ecosystems has been well-documented. Studies show that ZnO nanoparticles can induce oxidative stress, leading to tissue damage, immune responses, and developmental abnormalities in aquatic organisms [121]. Additionally, the bioaccumulation of ZnO NPs within organisms is influenced by nanoparticle concentration, exposure duration, and particle size [122]. Smaller nanoparticles are more readily absorbed by organisms, resulting in higher bioaccumulation and toxicity. These factors must be considered when evaluating the potential environmental impacts of ZnO nanoparticles.
Although ZnO NPs may offer benefits in terms of livestock growth and antimicrobial activity, careful management of their environmental impact is essential to prevent the accumulation of toxic zinc in soil and water. Future research should focus on the environmental behavior of ZnO NPs, particularly their transformation processes and bioaccumulation, in order to develop sustainable strategies for their use in animal feed.

7. Regulatory Landscape and Mitigation Strategies

Current regulatory frameworks for zinc supplementation in animal feed lack specific provisions addressing the unique properties of zinc oxide nanoparticles (ZnO NPs), despite evidence of their distinct ecotoxicological profiles compared to conventional zinc compounds. Specific regulatory standards are detailed in Table 3. To bridge this regulatory gap and mitigate risks associated with ZnO NPs, a multi-faceted strategy is essential. Dosage optimization must be prioritized, as studies indicate that high concentrations (>40 mg/kg) induce organ toxicity—evidenced by elevated serum creatinine, AST, ALT, and CK-MB levels in livestock—while lower doses sustain growth promotion and antimicrobial efficacy without adverse effects. Combining ZnO NPs with conventional zinc sources further reduces nephrotoxicity and hepatotoxicity risks. Long-term safety assessments should integrate advanced omics approaches (transcriptomics, proteomics, and metabolomics) to identify toxicity biomarkers, such as dysregulation of the AMPK-ULK1 pathway and NCOA4-mediated ferritin autophagy, which drive ferroptosis and vascular endothelial damage. Concurrently, environmental impact mitigation requires closed-loop waste management systems to recover excreted nanoparticles via membrane filtration, preventing aquatic ecosystem disruption, as ZnO NPs exhibit size-dependent bioaccumulation and induce oxidative stress in organisms like zebrafish. Standardization of green synthesis protocols is critical to minimize batch variability; physicochemical characterization (size: ±5 nm, zeta potential: ±10 mV) and surface functionalization using plant-derived stabilizers (e.g., mulberry extracts) enhance nanoparticle stability while reducing ecotoxicity [10,110]. Regulatory bodies must enforce traceability systems (e.g., isotopic labeling with 68Zn) and real-time monitoring technologies to align ZnO NP applications with One Health principles, ensuring sustainable adoption in global animal production systems [114].

8. Summary and Future Prospects

Zinc oxide nanoparticles (ZnO NPs) have emerged as a promising alternative to traditional feed additives in animal nutrition, offering significant antimicrobial, growth-promoting, and immune-regulatory benefits. Their unique physicochemical properties, such as nanoscale size and high surface area, contribute to their effectiveness in improving animal health and productivity. ZnO NPs have shown strong antibacterial activity, promoting growth and enhancing the immune response in livestock, thus providing a viable solution to the growing concerns over antibiotic resistance in animal production.
Furthermore, ZnO NPs have demonstrated their potential in improving meat, egg, and milk quality, as well as promoting better nutrient absorption and intestinal health. The green synthesis methods used for ZnO NP production have also garnered attention, as they provide an environmentally friendly approach with reduced toxicity, thereby contributing to more sustainable agricultural practices.
However, despite the promising applications of ZnO NPs in animal feed, several key challenges remain. First, the optimization of their dosage is critical to ensure that the benefits of ZnO NPs are maximized without causing adverse effects due to overuse. Additionally, long-term safety studies are still lacking, particularly regarding potential risks to animal health with prolonged exposure to ZnO NPs. Furthermore, the ecological toxicity and environmental behavior of ZnO NPs have not been fully explored, and more research is needed to understand their fate and impact on the environment.
Looking ahead, ZnO NPs hold great promise for revolutionizing animal nutrition and supporting sustainable livestock production. However, future research should focus on optimizing their dosage, enhancing bioavailability, and investigating the mechanisms underlying their antimicrobial efficacy. Moreover, understanding the environmental implications and ensuring the safe, sustainable use of ZnO NPs in animal feed will be essential for their continued application in the livestock industry. Developing safe and effective regulatory frameworks will be crucial to ensure their responsible use and mitigate any potential ecological risks.

Author Contributions

Writing—draft, J.Y.; writing—review and editing, D.X. and M.L.; supervision, M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (grant no. 32273074, grant no. 31972746, grant no. 31772809).

Data Availability Statement

Data generated in this study are presented in the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Behrens, I.; Pena, A.I.V.; Alonso, M.J.; Kissel, T. Comparative uptake studies of bioadhesive and non-bioadhesive nanoparticles in human intestinal cell lines and rats: The effect of mucus on particle adsorption and transport. Pharm. Res. 2002, 19, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
  2. Lee, K.P.; Kelly, D.P.; Schneider, P.W.; Trochimowicz, H.J. Inhalation toxicity study on rats exposed to titanium tetrachloride atmospheric hydrolysis products for two years. Toxicol. Appl. Pharmacol. 1986, 83, 30–45. [Google Scholar] [CrossRef] [PubMed]
  3. Oberdorster, G.; Ferin, J.; Lehnert, B.E. Correlation between particle size, in vivo particle persistence, and lung injury. Environ. Health Perspect. 1994, 102, 173–179. [Google Scholar] [CrossRef]
  4. Jamdagni, P.; Khatri, P.; Rana, J.S. Green synthesis of zinc oxide nanoparticles using flower extract of Nyctanthes arbor-tristis and their antifungal activity. J. King Saud Univ. Sci. 2018, 30, 168–175. [Google Scholar] [CrossRef]
  5. Sirelkhatim, A.; Mahmud, S.; Seeni, A.; Kaus, N.H.M.; Ann, L.C.; Bakhori, S.K.M.; Hasan, H.; Mohamad, D. Review on zinc oxide nanoparticles: Antibacterial activity and toxicity mechanism. Nano Micro Lett. 2015, 7, 219–242. [Google Scholar] [CrossRef]
  6. Banumathi, B.; Vaseeharan, B.; Ishwarya, R.; Govindarajan, M.; Alharbi, N.S.; Kadaikunnan, S.; Khaled, J.M.; Benelli, G. Toxicity of herbal extracts used in ethno-veterinary medicine and green-encapsulated ZnO nanoparticles against Aedes aegypti and microbial pathogens. Parasitol. Res. 2017, 116, 1637–1651. [Google Scholar] [CrossRef] [PubMed]
  7. Bogdan, J.; Plawinska-Czarnak, J.; Zarzynska, J. Nanoparticles of titanium and zinc oxides as novel agents in tumor treatment: A review. Nanoscale Res. Lett. 2017, 12, 225. [Google Scholar] [CrossRef]
  8. Al-Khaial, M.Q.; Chan, S.Y.; Abu-Zurayk, R.A.; Alnairat, N. Biosynthesis and characterization of zinc oxide nanoparticles (ZnO-NPs) utilizing banana peel extract. Inorganics 2024, 12, 121. [Google Scholar] [CrossRef]
  9. Gangadoo, S.; Stanley, D.; Hughes, R.J.; Moore, R.J.; Chapman, J. Nanoparticles in feed: Progress and prospects in poultry research. Trends Food Sci. Technol. 2016, 58, 115–126. [Google Scholar] [CrossRef]
  10. Ghanbari, M.; Bazarganipour, M.; Salavati-Niasari, M. Photodegradation and removal of organic dyes using cui nanostructures, green synthesis and characterization. Sep. Purif. Technol. 2017, 173, 27–36. [Google Scholar] [CrossRef]
  11. Kwabena, D.E.; Wee, B.S.; Fun, C.S.; Kok, K.Y.; Assim, Z.B.; Aquisman, A.E. Comparative evaluation of antibacterial efficacy of biological synthesis of ZnO nanoparticles using fresh leaf extract and fresh stem-bark of carica papaya. Nano Biomed. Eng. 2019, 11, 264–271. [Google Scholar] [CrossRef]
  12. Suhag, D.; Thakur, P.; Thakur, A. Introduction to Nanotechnology. In Integrated Nanomaterials and Their Applications; Suhag, D., Thakur, A., Thakur, P., Eds.; Springer Nature: Singapore, 2023; pp. 1–17. [Google Scholar]
  13. Dey, S.; Mohanty, D.l.; Divya, N.; Bakshi, V.; Mohanty, A.; Rath, D.; Das, S.; Mondal, A.; Roy, S.; Sabui, R. A critical review on zinc oxide nanoparticles: Synthesis, properties and biomedical applications. Intell. Pharm. 2024, 3, 53–70. [Google Scholar] [CrossRef]
  14. Li, Y.; Li, J.; Li, M.; Sun, J.; Shang, X.; Ma, Y. Biological mechanism of ZnO nanomaterials. J. Appl. Toxicol. 2024, 44, 107–117. [Google Scholar] [CrossRef]
  15. Mandal, A.K.; Behera, A.K.; Parida, S.; Nayak, R.; Hembram, P.; Behera, C.; Lone, S.A.; Davoodbasha, M.; Jena, M. Microalgal nanofactory: Green synthesis of zinc oxide nanoparticles and their antibacterial and antioxidant potentials. BioNanoScience 2024, 15, 119. [Google Scholar] [CrossRef]
  16. Rosi, N.L.; Mirkin, C.A. Nanostructures in biodiagnostics. Chem. Rev. 2005, 105, 1547–1562. [Google Scholar] [CrossRef] [PubMed]
  17. Feng, Q.L.; Wu, J.; Chen, G.Q.; Cui, F.Z.; Kim, T.N.; Kim, J.O. A mechanistic study of the antibacterial effect of silver ions on Escherichia coli and Staphylococcus aureus. J. Biomed. Mater. Res. 2000, 52, 662–668. [Google Scholar] [CrossRef]
  18. Mendes, C.R.; Dilarri, G.; Forsan, C.F.; Sapata, V.d.M.R.; Lopes, P.R.M.; de Moraes, P.B.; Montagnolli, R.N.; Ferreira, H.; Bidoia, E.D. Antibacterial action and target mechanisms of zinc oxide nanoparticles against bacterial pathogens. Sci. Rep. 2022, 12, 2658. [Google Scholar] [CrossRef] [PubMed]
  19. Adams, L.K.; Lyon, D.Y.; McIntosh, A.; Alvarez, P.J.J. Comparative toxicity of nano-scale TiO2, SiO2 and ZnO water suspensions. Water Sci. Technol. 2006, 54, 327–334. [Google Scholar] [CrossRef]
  20. Lipovsky, A.; Nitzan, Y.; Gedanken, A.; Lubart, R. Antifungal activity of ZnO nanoparticles—The role of ROS mediated cell injury. Nanotechnology 2011, 22, 105101. [Google Scholar] [CrossRef]
  21. Sawai, J.; Shoji, S.; Igarashi, H.; Hashimoto, A.; Kokugan, T.; Shimizu, M.; Kojima, H. Hydrogen peroxide as an antibacterial factor in zinc oxide powder slurry. J. Ferment. Bioeng. 1998, 86, 521–522. [Google Scholar] [CrossRef]
  22. Li, M.; Zhu, L.; Lin, D. Toxicity of ZnO nanoparticles to Escherichia coli: Mechanism and the influence of medium components. Environ. Sci. Technol. 2011, 45, 1977–1983. [Google Scholar] [CrossRef] [PubMed]
  23. Amro, N.A.; Kotra, L.P.; Wadu-Mesthrige, K.; Bulychev, A.; Mobashery, S.; Liu, G.-y. High-resolution atomic force microscopy studies of the Escherichia coli outer membrane:  structural basis for permeability. Langmuir 2000, 16, 2789–2796. [Google Scholar] [CrossRef]
  24. Divya, M.; Chen, J.; Durán-Lara, E.F.; Kim, K.-s.; Vijayakumar, S. Revolutionizing healthcare: Harnessing nano biotechnology with zinc oxide nanoparticles to combat biofilm and bacterial infections-A short review. Microb. Pathogen. 2024, 191, 106679. [Google Scholar] [CrossRef]
  25. Lok, C.N.; Ho, C.M.; Chen, R.; He, Q.Y.; Yu, W.Y.; Sun, H.; Tam, P.K.; Chiu, J.F.; Che, C.M. Proteomic analysis of the mode of antibacterial action of silver nanoparticles. J. Proteome Res. 2006, 5, 916–924. [Google Scholar] [CrossRef]
  26. Meruvu, H.; Vangalapati, M.; Chaitanya, S.; Bammidi, S.R. Synthesis and characterization of zinc oxide nanoparticles and its antimicrobial activity against bacillus subtilis and Escherichia coli. J. Rasayan. Chem. 2011, 4, 217–222. [Google Scholar]
  27. Nirmala, M.; Nair, M.G.; Rekha, K.; Anukaliani, A.; Samdarshi, S.; Nair, R.G. Photocatalytic activity of zno nanopowders synthesized by DC thermal plasma. Afr. J. Basic Appl. Sci. 2010, 2, 161–166. [Google Scholar]
  28. Zhang, H.; Chen, B.; Jiang, H.; Wang, C.; Wang, H.; Wang, X. A strategy for ZnO nanorod mediated multi-mode cancer treatment. Biomaterials 2011, 32, 1906–1914. [Google Scholar] [CrossRef]
  29. Gharpure, S.; Ankamwar, B. Synthesis and antimicrobial properties of zinc oxide nanoparticles. J. Nanosci. Nanotechnol. 2020, 20, 5977–5996. [Google Scholar] [CrossRef] [PubMed]
  30. 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. [Google Scholar] [CrossRef]
  31. Agarwal, H.; Nakara, A.; Shanmugam, V.K. Anti-inflammatory mechanism of various metal and metal oxide nanoparticles synthesized using plant extracts: A review. Biomed. Pharmacother. 2019, 109, 2561–2572. [Google Scholar] [CrossRef]
  32. Agarwal, H.; Shanmugam, V. A review on anti-inflammatory activity of green synthesized zinc oxide nanoparticle: Mechanism-based approach. Bioorg. Chem. 2020, 94, 103423. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, M.H.; Jeong, H.J. Zinc oxide nanoparticles suppress LPS-Induced NF-κB activation by inducing A20, a negative regulator of NF-κB, in RAW 264.7 macrophages. J. Nanosci. Nanotechnol. 2015, 15, 6509–6515. [Google Scholar] [CrossRef] [PubMed]
  34. Britt, R.D., Jr.; Locy, M.L.; Tipple, T.E.; Nelin, L.D.; Rogers, L.K. Lipopolysaccharide-induced cyclooxygenase-2 expression in mouse transformed clara cells. Cell. Physiol. Biochem. 2012, 29, 213–222. [Google Scholar] [CrossRef] [PubMed]
  35. Suschek, C.V.; Schnorr, O.; Kolb-Bachofen, V. The role of iNOS in chronic inflammatory processes in vivo: Is it damage-promoting, protective, or active at all? Curr. Mol. Med. 2004, 4, 763–775. [Google Scholar] [CrossRef]
  36. Nagajyothi, P.C.; Cha, S.J.; Yang, I.J.; Sreekanth, T.V.M.; Kim, K.J.; Shin, H.M. Antioxidant and anti-inflammatory activities of zinc oxide nanoparticles synthesized using Polygala tenuifolia root extract. J. Photochem. Photobiol. B Biol. 2015, 146, 10–17. [Google Scholar] [CrossRef]
  37. Das, D.; Nath, B.C.; Phukon, P.; Kalita, A.; Dolui, S.K. Synthesis of ZnO nanoparticles and evaluation of antioxidant and cytotoxic activity. Colloids Surf. B Biointerfaces 2013, 111, 556–560. [Google Scholar] [CrossRef]
  38. Loganathan, S.; Shivakumar, M.S.; Karthi, S.; Nathan, S.S.; Selvam, K. Metal oxide nanoparticle synthesis (ZnO-NPs) of Knoxia sumatrensis (Retz.) DC. Aqueous leaf extract and It’s evaluation of their antioxidant, anti-proliferative and larvicidal activities. Toxicol. Rep. 2021, 8, 64–72. [Google Scholar] [CrossRef]
  39. Moghaddam, A.B.; Moniri, M.; Azizi, S.; Rahim, R.A.; Ariff, A.B.; Saad, W.Z.; Namvar, F.; Navaderi, M. Biosynthesis of ZnO Nanoparticles by a New Pichia kudriavzevii Yeast Strain and Evaluation of Their Antimicrobial and Antioxidant Activities. Molecules 2017, 22, 872. [Google Scholar] [CrossRef]
  40. Sohail, M.F.; Rehman, M.; Hussain, S.Z.; Huma, Z.-E.; Shahnaz, G.; Qureshi, O.S.; Khalid, Q.; Mirza, S.; Hussain, I.; Webster, T.J. Green synthesis of zinc oxide nanoparticles by Neem extract as multi-facet therapeutic agents. J. Drug Deliv. Sci. Technol. 2020, 59, 101911. [Google Scholar] [CrossRef]
  41. Singh, T.A.; Sharma, A.; Tejwan, N.; Ghosh, N.; Das, J.; Sil, P.C. A state of the art review on the synthesis, antibacterial, antioxidant, antidiabetic and tissue regeneration activities of zinc oxide nanoparticles. Adv. Colloid Interface Sci. 2021, 295, 102495. [Google Scholar] [CrossRef]
  42. Rabiee, N.; Bagherzadeh, M.; Kiani, M.; Ghadiri, A.M.; Etessamifar, F.; Jaberizadeh, A.H.; Shakeri, A. Biosynthesis of copper oxide nanoparticles with potential biomedical applications. Int. J. Nanomed. 2020, 15, 3983–3999. [Google Scholar] [CrossRef]
  43. Kalaimurugan, D.; Lalitha, K.; Durairaj, K.; Sivasankar, P.; Park, S.; Nithya, K.; Shivakumar, M.S.; Liu, W.-C.; Balamuralikrishnan, B.; Venkatesan, S. Biogenic synthesis of ZnO nanoparticles mediated from Borassus flabellifer (Linn): Antioxidant, antimicrobial activity against clinical pathogens, and photocatalytic degradation activity with molecular modeling. Environ. Sci. Pollut. Res. 2022, 29, 86308–86319. [Google Scholar] [CrossRef] [PubMed]
  44. Jan, H.; Shah, M.; Andleeb, A.; Faisal, S.; Khattak, A.; Rizwan, M.; Drouet, S.; Hano, C.; Abbasi, B.H. Plant-based synthesis of zinc oxide nanoparticles (ZnO-NPs) using aqueous leaf extract of Aquilegia pubiflora their Antiproliferative activity against HepG2 cells inducing reactive oxygen species and other In Vitro properties. Oxid. Med. Cell Longev. 2021, 2021, 4786227. [Google Scholar] [CrossRef] [PubMed]
  45. Asif, N.; Fatima, S.; Aziz, M.N.; Shehzadi; Zaki, A.; Fatma, T. Biofabrication and characterization of cyanobacteria derived ZnO NPs for their bioactivity comparison with commercial chemically synthesized nanoparticles. Bioorg. Chem. 2021, 113, 104999. [Google Scholar] [CrossRef]
  46. Kiyani, M.M.; Butt, M.A.; Rehman, H.; Ali, H.; Hussain, S.A.; Obaid, S.; Arif Hussain, M.; Mahmood, T.; Bokhari, S.A.I. Antioxidant and anti-gout effects of orally administered zinc oxide nanoparticles in gouty mice. J. Trace Elem. Med. Biol. 2019, 56, 169–177. [Google Scholar] [CrossRef] [PubMed]
  47. Hafez, A.; Nassef, E.; Fahmy, M.; Elsabagh, M.; Bakr, A.; Hegazi, E. Impact of dietary nano-zinc oxide on immune response and antioxidant defense of broiler chickens. Environ. Sci. Pollut. Res. Int. 2020, 27, 19108–19114. [Google Scholar] [CrossRef]
  48. Elbahr, A. Kaff Al-‘AwÂm: Saat Kiai Hasyim Berbicara Sarekat Islam. Int. J. Pegon. Islam Nusantara Civ. 2020, 3, 1–79. [Google Scholar] [CrossRef]
  49. Wang, C.; Zhang, L.; Ying, Z.; He, J.; Zhou, L.; Zhang, L.; Zhong, X.; Wang, T. Effects of dietary zinc oxide nanoparticles on growth, diarrhea, mineral deposition, intestinal morphology, and barrier of weaned piglets. Biol. Trace Elem. Res. 2018, 185, 364–374. [Google Scholar] [CrossRef]
  50. Ramiah, S.K.; Awad, E.A.; Mookiah, S.; Idrus, Z. Effects of zinc oxide nanoparticles on growth performance and concentrations of malondialdehyde, zinc in tissues, and corticosterone in broiler chickens under heat stress conditions. Poult. Sci. 2019, 98, 3828–3838. [Google Scholar] [CrossRef]
  51. Yaqub, A.; Nasir, M.; Kamran, M.; Majeed, I.; Arif, A. Immunomodulation, fish health and resistance to Staphylococcus aureus of nile tilapia (Oreochromis niloticus) fed diet supplemented with zinc oxide nanoparticles and zinc acetate. Biol. Trace Elem. Res. 2023, 201, 4912–4925. [Google Scholar] [CrossRef]
  52. Amin, Y.A.; Abdelaziz, S.G.; Said, A.H. Treatment of postpartum endometritis induced by multidrug-resistant bacterial infection in dairy cattle by green synthesized zinc oxide nanoparticles and in vivo evaluation of its broad spectrum antimicrobial activity in cow uteri. Res. Vet. Sci. 2023, 165, 105074. [Google Scholar] [CrossRef] [PubMed]
  53. Abbasi, M.; Dastar, B.; Afzali, N.; Shargh, M.S.; Hashemi, S.R. The effects of nano and micro particle size of zinc oxide on performance, fertility, hatchability, and egg quality characteristics in laying Japanese quail. Biol. Trace Elem. Res. 2022, 200, 2338–2348. [Google Scholar] [CrossRef] [PubMed]
  54. Khafajah, Y.; Shaheen, M.; Natour, D.E.; Merheb, M.; Matar, R.; Borjac, J. Neuroprotective effects of zinc oxide nanoparticles in a rotenone-induced mouse model of parkinson’s disease. Nanotheranostics 2024, 8, 497–505. [Google Scholar] [CrossRef] [PubMed]
  55. Attia, F.M.; Kassab, R.B.; Ahmed-Farid, O.A.; Abdel Moneim, A.E.; El-Yamany, N.A. Zinc oxide nanoparticles attenuated neurochemical and histopathological alterations associated with aluminium chloride intoxication in rats. Biol. Trace Elem. Res. 2025, 203, 2058–2071. [Google Scholar] [CrossRef]
  56. Petrič, D.; Mikulová, K.; Bombárová, A.; Batťányi, D.; Čobanová, K.; Kopel, P.; Łukomska, A.; Pawlak, P.; Sidoruk, P.; Kotwica, S.; et al. Efficacy of zinc nanoparticle supplementation on ruminal environment in lambs. BMC Vet. Res. 2024, 20, 425. [Google Scholar] [CrossRef]
  57. Ismail, H.; El-Araby, I.E. Effect of dietary zinc oxide nanoparticles supplementation on biochemical, hematological and genotoxicity parameters in rabbits. Int. J. Curr. Adv. Res. 2018, 6, 2108–2115. [Google Scholar]
  58. Hussain, Z.; Khan, J.A.; Anwar, H.; Andleeb, N.; Murtaza, S.; Ashar, A.; Arif, I. Synthesis, characterization, and pharmacological evaluation of zinc oxide nanoparticles formulation. Toxicol. Ind. Health 2018, 34, 753–763. [Google Scholar] [CrossRef]
  59. Safaei-Ghomi, J.; Ghasemzadeh, M.A.; Zahedi, S. ZnO nanoparticles: A highly effective and readily recyclable catalyst for the one-pot synthesis of 1,8-dioxo-decahydroacridine and 1,8-dioxooctahydro-xanthene derivatives. J. Mex. Chem. Soc. 2017, 57, 1–7. [Google Scholar] [CrossRef]
  60. Liu, Y.; He, L.; Mustapha, A.; Li, H.; Hu, Z.Q.; Lin, M. Antibacterial activities of zinc oxide nanoparticles against Escherichia coli O157:H7. J. Appl. Microbiol. 2009, 107, 1193–1201. [Google Scholar] [CrossRef]
  61. Raghupathi, K.R.; Koodali, R.T.; Manna, A.C. Size-dependent bacterial growth inhibition and mechanism of antibacterial activity of zinc oxide nanoparticles. Langmuir 2011, 27, 4020–4028. [Google Scholar] [CrossRef]
  62. Douglas, L.J. Candida biofilms and their role in infection. Trends Microbiol. 2003, 11, 30–36. [Google Scholar] [CrossRef] [PubMed]
  63. Yamamoto, O.; Iida, Y. Antifungal Characteristics of Spherical Carbon Materials with Zinc Oxide. J. Ceramic. Soc. Jpn. 2003, 111, 614–616. [Google Scholar] [CrossRef]
  64. Fozouni, L.; Tahaei, M. Anticandidal effects of zinc oxide nanoparticles on fluconazole-resistant candida isolates causing diarrhea in calves, in vitro. Arch. Razi Inst. 2023, 78, 499–504. [Google Scholar] [CrossRef] [PubMed]
  65. Awad, N.F.S.; Hashem, Y.M.; Elshater, N.S.; Khalifa, E.; Hamed, R.I.; Nossieur, H.H.; Abd-Allah, E.M.; Elazab, S.T.; Nassan, M.A.; El-Hamid, M.I.A. Therapeutic potentials of aivlosin and/or zinc oxide nanoparticles against Mycoplasma gallisepticum and/or Ornithobacterium rhinotracheale with a special reference to the effect of zinc oxide nanoparticles on aivlosin tissue residues: An in vivo approach. Poult. Sci. 2022, 101, 101884. [Google Scholar] [CrossRef]
  66. El-Kady, A.M.; Hassan, A.S.; Mohamed, K.; Alfaifi, M.S.; Elshazly, H.; Alamri, Z.Z.; Wakid, M.H.; Gattan, H.S.; Altwaim, S.A.; Al-Megrin, W.A.I.; et al. Zinc oxide nanoparticles produced by Zingiber officinale ameliorates acute toxoplasmosis-induced pathological and biochemical alterations and reduced parasite burden in mice model. PLoS Neglected Trop. Dis. 2023, 17, e0011447. [Google Scholar] [CrossRef]
  67. Lail, N.U.; Sattar, A.; Omer, M.O.; Hafeez, M.A.; Khalid, A.R.; Mahmood, S.; Shabbir, M.A.; Ahmed, W.; Aleem, M.T.; Alouffi, A.; et al. Biosynthesis and characterization of zinc oxide nanoparticles using Nigella sativa against coccidiosis in commercial poultry. Sci. Rep. 2023, 13, 6568. [Google Scholar] [CrossRef]
  68. Gammoh, N.Z.; Rink, L. Zinc in Infection and Inflammation. Nutrients 2017, 9, 624. [Google Scholar] [CrossRef]
  69. Fraker, P.J.; DePasquale-Jardieu, P.; Zwickl, C.M.; Luecke, R.W. Regeneration of T-cell helper function in zinc-deficient adult mice. Proc. Natl. Acad. Sci. USA 1978, 75, 5660–5664. [Google Scholar] [CrossRef] [PubMed]
  70. Kong, T.; Zhang, S.H.; Zhang, C.; Zhang, J.L.; Yang, F.; Wang, G.Y.; Yang, Z.J.; Bai, D.Y.; Shi, Y.Y.; Liu, T.Q.; et al. The Effects of 50 nm unmodified Nano-ZnO on lipid metabolism and semen quality in male mice. Biol. Trace Elem. Res. 2019, 194, 432–442. [Google Scholar] [CrossRef]
  71. Liu, H.; Bai, M.; Xu, K.; Zhou, J.; Zhang, X.; Yu, R.; Huang, R.; Yin, Y. Effects of different concentrations of coated nano zinc oxide material on fecal bacterial composition and intestinal barrier in weaned piglets. J. Sci. Food Agric. 2021, 101, 735–745. [Google Scholar] [CrossRef]
  72. Trckova, M.; Lorencova, A.; Hazova, K.; Zajacova, Z. Prophylaxis of post-weaning diarrhoea in piglets by zinc oxide and sodium humate. Vet. Med. 2015, 60, 351–360. [Google Scholar] [CrossRef]
  73. Ahmadi, F.; Ebrahimnezhad, Y.; Sis, N.M.; Ghalehkandi, J.G. The effects of zinc oxide nanoparticles on performance, digestive organs and serum lipid concentrations in broiler chickens during starter period. Int. J. Biosci. 2013, 3, 23–29. [Google Scholar] [CrossRef]
  74. Jin, T.; Sun, D.; Su, J.Y.; Zhang, H.; Sue, H.J. Antimicrobial efficacy of zinc oxide quantum dots against Listeria monocytogenes, Salmonella Enteritidis, and Escherichia coli O157:H7. J. Food Sci. 2009, 74, M46–M52. [Google Scholar] [CrossRef] [PubMed]
  75. Milani, N.C.; Sbardella, M.; Ikeda, N.Y.; Arno, A.; Mascarenhas, B.C.; Miyada, V.S. Dietary zinc oxide nanoparticles as growth promoter for weanling pigs. Anim. Feed. Sci. Technol. 2017, 227, 13–23. [Google Scholar] [CrossRef]
  76. Sagar, P.; Mandal, A.B.; Akbar, N.; Dinani, O.P. Effect of different levels and sources of zinc on growth performance and immunity of broiler chicken during summer. Int. J. Curr. Microbiol. App. Sci. 2018, 7, 459–471. [Google Scholar] [CrossRef]
  77. Qu, J.; Zuo, X.; Xu, Q.; Li, M.; Zou, L.; Tao, R.; Liu, X.; Wang, X.; Wang, J.; Wen, L.; et al. Effect of two particle sizes of nano zinc oxide on growth performance, immune function, digestive tract morphology, and intestinal microbiota composition in broilers. Animals 2023, 13, 1454. [Google Scholar] [CrossRef] [PubMed]
  78. Ramiah, S.K.; Atta Awad, E.; Hemly, N.I.M.; Ebrahimi, M.; Joshua, O.; Jamshed, M.; Saminathan, M.; Soleimani, A.F.; Idrus, Z. Effects of zinc oxide nanoparticles on regulatory appetite and heat stress protein genes in broiler chickens subjected to heat stress. J. Anim. Sci. 2020, 98, skaa300. [Google Scholar] [CrossRef]
  79. Abdel-Latif, H.M.R.; Abdel-Tawwab, M.; Khafaga, A.F.; Dawood, M.A.O. Dietary oregano essential oil improved antioxidative status, immune-related genes, and resistance of common carp (Cyprinus carpio L.) to Aeromonas hydrophila infection. Fish Shellfish Immunol. 2020, 104, 1–7. [Google Scholar] [CrossRef]
  80. Sallam, A.E.; Mansour, A.T.; Alsaqufi, A.S.; El-Sayed Salem, M.; El-Feky, M.M. Growth performance, anti-oxidative status, innate immunity, and ammonia stress resistance of Siganus rivulatus fed diet supplemented with zinc and zinc nanoparticles. Aquac. Rep. 2020, 18, 100410. [Google Scholar] [CrossRef]
  81. Swain, P.S.; Rao, S.B.N.; Rajendran, D.; Dominic, G.; Selvaraju, S. Nano zinc, an alternative to conventional zinc as animal feed supplement: A review. Anim. Nutr. 2016, 2, 134–141. [Google Scholar] [CrossRef]
  82. Awad, A.; Zaglool, A.W.; Ahmed, S.A.A.; Khalil, S.R. Transcriptomic profile change, immunological response and disease resistance of Oreochromis niloticus fed with conventional and Nano-Zinc oxide dietary supplements. Fish Shellfish Immunol. 2019, 93, 336–343. [Google Scholar] [CrossRef] [PubMed]
  83. Mahmoud, H.K.; Al-Sagheer, A.A.; Reda, F.M.; Mahgoub, S.A.; Ayyat, M.S. Dietary curcumin supplement influence on growth, immunity, antioxidant status, and resistance to Aeromonas hydrophila in Oreochromis niloticus. Aquaculture 2017, 475, 16–23. [Google Scholar] [CrossRef]
  84. Sherif, A.H.; Abdelsalam, M.; Ali, N.G.; Mahrous, K.F. Zinc oxide nanoparticles boost the immune responses in oreochromis niloticus and improve disease resistance to aeromonas hydrophila infection. Biol. Trace Elem. Res. 2023, 201, 927–936. [Google Scholar] [CrossRef] [PubMed]
  85. Tawfik, M.; Moustafa, M.; Abumourad, I.; El-Meliegy, E.; Refai, M. Evaluation of Nano Zinc Oxide feed additive on tilapia Growth and Immunity. In Proceedings of the 15th International Conference on Environmental Science and Technology, Rhodes, Greece, 31 August–2 September 2017; pp. 1–9. [Google Scholar]
  86. Mishra, A.; Swain, R.; Mishra, S.; Panda, N.; Sethy, K. Growth performance and serum biochemical parameters as affected by nano zinc supplementation in layer chicks. Indian J. Anim. Nutr. 2014, 31, 384–388. [Google Scholar]
  87. Yang, Z.P.; Sun, L.P. Effects of nanometre ZnO on growth performance of early weaned piglets. J. Shanxi Agric. Sci. 2006, 3, 577–588. [Google Scholar] [CrossRef]
  88. Rajendran, D.; Kumar, G.; Ramakrishnan, S.; Shibi, T. Enhancing the milk production and immunity in Holstein Friesian crossbred cow by supplementing novel nano zinc oxide. Res. J. Biotechnol. 2013, 8, 11–17. [Google Scholar]
  89. Chen, J.; Wang, W.; Wang, Z. Effect of nano-zinc oxide supplementation on rumen fermentation in vitro. Chin. J. Anim. Nutr. 2011, 23, 1415–1421. [Google Scholar]
  90. Lönnerdal, B. Dietary factors influencing zinc absorption. J. Nutr. 2000, 130, 1378S–1383S. [Google Scholar] [CrossRef]
  91. Khajeh Bami, M.; Afsharmanesh, M.; Salarmoini, M.; Tavakoli, H. Effect of zinc oxide nanoparticles and Bacillus coagulans as probiotic on growth, histomorphology of intestine, and immune parameters in broiler chickens. Comp. Clin. Pathol. 2018, 27, 399–406. [Google Scholar] [CrossRef]
  92. Radi, A.M.; Abdel Azeem, N.M.; El-Nahass, E.S. Comparative effects of zinc oxide and zinc oxide nanoparticle as feed additives on growth, feed choice test, tissue residues, and histopathological changes in broiler chickens. Environ. Sci. Pollut. Res. Int. 2021, 28, 5158–5167. [Google Scholar] [CrossRef]
  93. Fawaz, M.A.; Südekum, K.H.; Hassan, H.A.; Abdel-Wareth, A.A.A. Productive, physiological and nutritional responses of laying hens fed different dietary levels of turmeric powder. J. Anim. Physiol. Anim. Nutr. 2023, 107, 214–221. [Google Scholar] [CrossRef]
  94. Abdel-Wareth, A.A.A.; Hussein, K.R.A.; Ismail, Z.S.H.; Lohakare, J. Effects of zinc oxide nanoparticles on the performance of broiler chickens under hot climatic conditions. Biol. Trace Elem. Res. 2022, 200, 5218–5225. [Google Scholar] [CrossRef]
  95. Karimi, A.; Hosseini, S.K.; Nemati, Z.; Sheikhlou, M.R. Effects of different zinc sources on productive performance and egg quality, blood parameters and immune response in Japanese layer quail. Res. Anim. Prod. 2018, 9, 27–35. [Google Scholar] [CrossRef]
  96. Abedini, M.; Shariatmadari, F.; Karimi Torshizi, M.A.; Ahmadi, H. Effects of zinc oxide nanoparticles on the egg quality, immune response, zinc retention, and blood parameters of laying hens in the late phase of production. J. Anim. Physiol. Anim. Nutr. 2018, 102, 736–745. [Google Scholar] [CrossRef] [PubMed]
  97. Mao, S.-Y.; Lien, T.-F. Effects of nanosized zinc oxide and γ-polyglutamic acid on eggshell quality and serum parameters of aged laying hens. Arch. Anim. Nutr. 2017, 71, 373–383. [Google Scholar] [CrossRef]
  98. Alkhtib, A.; Scholey, D.; Carter, N.; Cave, G.W.V.; Hanafy, B.I.; Kempster, S.R.J.; Mekapothula, S.; Roxborough, E.T.; Burton, E.J. Bioavailability of methionine-coated zinc nanoparticles as a dietary supplement leads to improved performance and bone strength in broiler chicken production. Animals 2020, 10, 1482. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, Z.H.; Lu, L.; Wang, R.L.; Lei, H.L.; Li, S.F.; Zhang, L.Y.; Luo, X.G. Effects of supplemental zinc source and level on antioxidant ability and fat metabolism-related enzymes of broilers. Poult. Sci. 2015, 94, 2686–2694. [Google Scholar] [CrossRef]
  100. Mozhiarasi, V.; Karunakaran, R.; Raja, P.; Radhakrishnan, L. Effects of zinc oxide nanoparticles supplementation on growth performance, meat quality and serum biochemical parameters in broiler chicks. Biol. Trace Elem. Res. 2024, 202, 1683–1698. [Google Scholar] [CrossRef]
  101. Hassan, S.; Sharif, M.; Mirza, M.A.; Rehman, M.S.U. Effect of dietary supplementation of zinc nanoparticles prepared by different green methods on egg production, egg quality, bone mineralization, and antioxidant capacity in caged layers. Biol. Trace Elem. Res. 2023, 201, 5794–5804. [Google Scholar] [CrossRef]
  102. Mahmoud, U.T.; Abdel-Mohsein, H.S.; Mahmoud, M.A.M.; Amen, O.A.; Hassan, R.I.M.; Abd-El-Malek, A.M.; Rageb, S.M.M.; Waly, H.S.A.; Othman, A.A.; Osman, M.A. Effect of zinc oxide nanoparticles on broilers’ performance and health status. Trop. Anim. Health Prod. 2020, 52, 2043–2054. [Google Scholar] [CrossRef]
  103. El-Katcha, M.I.Y.; Soltan, M.A.; Elbadry, M. Effect of dietary replacement of inorganic zinc by organic or nanoparticles sources on growth performance, immune response and intestinal histopathology of broiler chicken. Alex. J. Vet. Sci. 2017, 55, 129–145. [Google Scholar] [CrossRef]
  104. Khah, M.M.; Ahmadi, F.; Amanlou, H. Influence of dietary different levels of zinc oxide nano particles on the yield and quality carcass of broiler chickens during starter stage. Indian J. Anim. Sci. 2015, 85, 287–290. [Google Scholar] [CrossRef]
  105. Lina, T.; Fenghua, Z.; Hui-ying, R.; Jian-yang, J.; Wenli, L. Effects of nano-zinc oxide on antioxidant function in broilers. Chin. J. Anim. Nutr. 2009, 21, 534–539. [Google Scholar]
  106. Zhou, X.-Q.; Hayat, Z.; Zhang, D.-D.; Li, M.-Y.; Hu, S.; Wu, Q.; Cao, Y.-F.; Yuan, Y. Zinc oxide nanoparticles: Synthesis, characterization, modification, and applications in food and agriculture. Processes 2023, 11, 1193. [Google Scholar] [CrossRef]
  107. Emmanuela de Andrade Vieira, J.; de Oliveira Ferreira, R.; Marcel Dos Reis Sampaio, D.; Pereira da Costa Araújo, A.; Malafaia, G. An insight on the mutagenicity and cytotoxicity of zinc oxide nanoparticles in Gallus gallus domesticus (Phasianidae). Chemosphere 2019, 231, 10–19. [Google Scholar] [CrossRef] [PubMed]
  108. Cufadar, Y.; Göçmen, R.; Kanbur, G.; Yıldırım, B. Effects of dietary different levels of nano, organic and inorganic zinc sources on performance, eggshell quality, bone mechanical parameters and mineral contents of the tibia, liver, serum and excreta in laying hens. Biol. Trace Elem. Res. 2020, 193, 241–251. [Google Scholar] [CrossRef]
  109. Abedin, S.N.; Baruah, A.; Baruah, K.K.; Bora, A.; Dutta, D.J.; Kadirvel, G.; Katiyar, R.; Doley, S.; Das, S.; Khargharia, G.; et al. Zinc oxide and selenium nanoparticles can improve semen quality and heat shock protein expression in cryopreserved goat (Capra hircus) spermatozoa. J. Trace Elem. Med. Biol. 2023, 80, 127296. [Google Scholar] [CrossRef]
  110. Soltani, L.; Samereh, S.; Mohammadi, T. Effects of different concentrations of zinc oxide nanoparticles on the quality of ram cauda epididymal spermatozoa during storage at 4 °C. Reprod. Domest. Anim. 2022, 57, 864–875. [Google Scholar] [CrossRef]
  111. Khodaei-Motlagh, M.; Masoudi, R.; Karimi-Sabet, M.J.; Hatefi, A. Supplementation of sperm cooling medium with Zinc and Zinc oxide nanoparticles preserves rooster sperm quality and fertility potential. Theriogenology 2022, 183, 36–40. [Google Scholar] [CrossRef]
  112. Youn, S.-M.; Choi, S.-J. Food additive zinc oxide nanoparticles: Dissolution, interaction, fate, cytotoxicity, and oral toxicity. Int. J. Mol. Sci. 2022, 23, 6074. [Google Scholar] [CrossRef]
  113. Bae, S.-H.; Yu, J.; Lee, T.G.; Choi, S.-J. Protein food matrix–ZnO nanoparticle interactions affect protein conformation, but may not be biological responses. Int. J. Mol. Sci. 2018, 19, 3926. [Google Scholar] [CrossRef] [PubMed]
  114. Hall, A.G.; King, J.C. The molecular basis for zinc bioavailability. Int. J. Mol. Sci. 2023, 24, 6561. [Google Scholar] [CrossRef]
  115. Li, Y.; Vulpe, C.; Lammers, T.; Pallares, R.M. Assessing inorganic nanoparticle toxicity through omics approaches. Nanoscale 2024, 16, 15928–15945. [Google Scholar] [CrossRef] [PubMed]
  116. Abdelnour, S.A.; Alagawany, M.; Hashem, N.M.; Farag, M.R.; Alghamdi, E.S.; Hassan, F.U.; Bilal, R.M.; Elnesr, S.S.; Dawood, M.A.O.; Nagadi, S.A.; et al. Nanominerals: Fabrication methods, benefits and hazards, and their applications in ruminants with special reference to selenium and zinc nanoparticles. Animals 2021, 11, 1916. [Google Scholar] [CrossRef]
  117. Fernández-Bertólez, N.; Alba-González, A.; Touzani, A.; Ramos-Pan, L.; Méndez, J.; Reis, A.T.; Quelle-Regaldie, A.; Sánchez, L.; Folgueira, M.; Laffon, B.; et al. Toxicity of zinc oxide nanoparticles: Cellular and behavioural effects. Chemosphere 2024, 363, 142993. [Google Scholar] [CrossRef] [PubMed]
  118. Fujihara, J.; Nishimoto, N. Review of zinc oxide nanoparticles: Toxicokinetics, tissue distribution for various exposure routes, toxicological effects, toxicity mechanism in mammals, and an approach for toxicity reduction. Biol. Trace Elem. Res. 2024, 202, 9–23. [Google Scholar] [CrossRef]
  119. Qin, X.; Zhang, J.; Wang, B.; Xu, G.; Yang, X.; Zou, Z.; Yu, C. Ferritinophagy is involved in the zinc oxide nanoparticles-induced ferroptosis of vascular endothelial cells. Autophagy 2021, 17, 4266–4285. [Google Scholar] [CrossRef]
  120. Yung, M.; Mouneyrac, C.; Leung, K. Ecotoxicity of zinc oxide nanoparticles in the marine environment. In Encyclopedia of Nanotechnology; Springer: Dordrecht, The Netherlands, 2015; pp. 1–17. [Google Scholar] [CrossRef]
  121. Amin, N.; Erfan, M.A. Environmental fate and toxicity of zinc oxide nanoparticles in aquatic ecosystems: A comprehensive review. J. Nat. Sci. Rev. 2025, 3, 104–125. [Google Scholar] [CrossRef]
  122. Voss, L.; Saloga, P.E.J.; Stock, V.; Böhmert, L.; Braeuning, A.; Thünemann, A.F.; Lampen, A.; Sieg, H. Environmental impact of ZnO nanoparticles evaluated by in vitro simulated digestion. ACS Appl. Nano Mater. 2020, 3, 724–733. [Google Scholar] [CrossRef]
Figure 1. Green synthesis of nano-zinc oxide and its advantages.
Figure 1. Green synthesis of nano-zinc oxide and its advantages.
Nanomaterials 15 01030 g001
Figure 2. Schematic representation of the antibacterial mechanisms of ZnO nanoparticles against bacterial cells, illustrating key processes such as membrane disruption, Zn2+ ion uptake leading to cellular energy depletion and DNA damage, reactive oxygen species (ROS) generation, and photocatalytic ROS production under UV light.
Figure 2. Schematic representation of the antibacterial mechanisms of ZnO nanoparticles against bacterial cells, illustrating key processes such as membrane disruption, Zn2+ ion uptake leading to cellular energy depletion and DNA damage, reactive oxygen species (ROS) generation, and photocatalytic ROS production under UV light.
Nanomaterials 15 01030 g002
Figure 3. Key anti-inflammatory mechanisms mediated by ZnO nanoparticles, including NF-κB pathway modulation, mast cell regulation, and inflammatory enzyme inhibition. Black arrows indicate promotion or activation of cytokines/proteins, while red arrows indicate inhibition or suppression.
Figure 3. Key anti-inflammatory mechanisms mediated by ZnO nanoparticles, including NF-κB pathway modulation, mast cell regulation, and inflammatory enzyme inhibition. Black arrows indicate promotion or activation of cytokines/proteins, while red arrows indicate inhibition or suppression.
Nanomaterials 15 01030 g003
Figure 4. Key toxicity mechanisms of ZnO nanoparticles, including cellular dissolution, ROS generation, inflammatory activation, genetic alterations, and ferroptosis.
Figure 4. Key toxicity mechanisms of ZnO nanoparticles, including cellular dissolution, ROS generation, inflammatory activation, genetic alterations, and ferroptosis.
Nanomaterials 15 01030 g004
Table 1. Mechanisms of antibacterial, anti-inflammatory, and antioxidant actions of ZnO NPs.
Table 1. Mechanisms of antibacterial, anti-inflammatory, and antioxidant actions of ZnO NPs.
FunctionMechanismDescription
AntibacterialROS GenerationZnO NPs produce reactive oxygen species, damaging bacterial cell membranes and inhibiting bacterial growth.
Zn2+ Ion ReleaseZn2+ ions penetrate bacterial cells, disrupting metabolic functions and inhibiting protein and nucleic acid synthesis, ultimately causing cell death.
Particle Size EffectThe nanoscale size of ZnO NPs increases surface area, enhancing interaction with bacteria and boosting antibacterial activity.
Bacterial Membrane DisruptionZnO NPs disrupt bacterial membrane integrity, leading to the leakage of cell contents and cell death.
Anti-inflammatoryInhibition of Pro-Inflammatory FactorsZnO NPs inhibit the generation of pro-inflammatory factors (e.g., TNF-α and IL-6), reducing the inflammatory response.
Reduction of Tissue DamageBy reducing free radical production, ZnO NPs mitigate tissue damage caused by inflammation, protecting cells.
Blocking of Cytokine SignalingZnO NPs block pathways such as NF-κB, reducing the production of inflammatory cytokines and alleviating inflammation.
Reduction in Inflammatory Enzyme ActivityZnO NPs decrease the expression of enzymes such as COX-2 and iNOS, which are associated with the amplification of inflammation.
AntioxidantFree Radical ScavengingZnO NPs eliminate free radicals, reducing damage to cellular membranes and structures caused by oxidative stress.
Enhancement of Antioxidant Enzyme ActivityZnO NPs activate antioxidant enzymes (e.g., SOD, CAT, and GSH-Px), neutralizing free radicals and increasing cellular antioxidant capacity.
Reduction in Malondialdehyde (MDA) LevelsZnO NPs reduce oxidative stress, leading to lower MDA levels, mitigating lipid peroxidation, and cellular damage.
Strengthening Cellular Antioxidant DefenseZnO NPs enhance intracellular antioxidant defense mechanisms, protecting cells from protein, DNA, and lipid damage.
Table 2. Biological effects of zinc oxide nanoparticles in animal models.
Table 2. Biological effects of zinc oxide nanoparticles in animal models.
Animal SpeciesSample SizeTrial DurationRoute of AdministrationZnO Nanoparticle DosageObserved EffectsReferences
Pigsn = 414 daysDietary supplementation800 mg/kgImproved growth performance, reduced diarrhea incidence, enhanced intestinal barrier function, modulated immune responses, decreased zinc excretion, promoted intestinal morphology[49]
Broilersn = 642 daysDietary supplementation40/60/100 mg/kgModulated appetite-related genes (ghrelin and CCK), regulated heat stress protein genes (HSP70 and HSP90)[50]
Oreochromis niloticusn = 158 weeksDietary supplementation20/40/60 mg/kgEnhanced antioxidant capacity, improved blood health, boosted immune function, optimized metabolic profiles, reduced liver damage, enhanced resistance to Staphylococcus aureus at 40 mg/kg dietary supplementation[51]
Dairy CattleTotal = 5005 days treatment + 45–60 days observationIntrauterine infusion20 μg/mL (150 mL daily, total 3 mg/day per cow)Effective clinical cure of multidrug-resistant uterine infections, induction of estrus and pregnancy, broad-spectrum antibacterial activity[52]
Quailsn = 428 daysDietary supplementation0–130 mg/kgImproved eggshell thickness, enhanced fertility, increased egg weight and eggshell surface, optimized egg production with reduced zinc requirement[53]
Micen = 628 daysIntraperitoneal injection5.6 mg/kgSignificantly increased dopamine levels in the brains of Parkinson’s disease mice, activated the expression of the dopa decarboxylase gene while maintaining tyrosine hydroxylase activity. It had no significant effect on norepinephrine or epinephrine levels, effectively improving motor dysfunction and reversing weight loss symptoms induced by rotenone[54]
Ratsn = 742 daysIntraperitoneal injection4 mg/kgAttenuated oxidative damage, suppressed neuroinflammatory response, inhibited neuronal apoptosis, restored neurotransmitter balance, mitigated histopathological alterations[55]
LambsSTE: n = 9/group
LTE: n = 7/group
42 daysDietary supplementationSTE: 80 mg Zn/kg diet (ZnO-NPs, ZnP-NPs)
LTE: 40 mg/kg (ZnO-NP40), 80 mg/kg (ZnO-NP80, ZnO-80)
In vitro: enhanced dry matter digestibility; short-term: reduced total bacterial population with ZnP-NPs; long-term: decreased ammonia-N concentration (ZnO-NP80), increased carboxymethyl cellulase and xylanase activities; histopathology: inflammatory changes in ruminal papillae and epithelium regardless of Zn form/dose/duration, improved feed-use efficiency via altered fermentation patterns enhancing SCFA transport capacity[56]
Rabbitsn = 1545 daysDietary supplementationGp.3: 60 mg/kg ZnO NPs
Gp.4: 30 mg/kg ZnO + 30 mg/kg ZnO NPs
Gp.3 (ZnO NPs): elevated liver enzymes (ALT/AST), hypoproteinemia/hypoalbuminemia, reduced lipid profile (TG/TC/HDL-C), increased renal markers (creatinine/urea), hepatic/renal oxidative stress (↑ MDA, ↓ CAT), leukocytosis/lymphocytosis, hepatic/renal DNA damage, severe hepatic hydropic degeneration and renal interstitial nephritis; Gp.4 (combined): mitigated adverse effects (less severe biochemical/oxidative/genotoxic changes and histopathology) while retaining zinc benefits[57]
Table 3. Regulatory standards for ZnO NPs vs. zinc compounds.
Table 3. Regulatory standards for ZnO NPs vs. zinc compounds.
Regulatory BodyStandardNP-Specific Clause
EFSA (EU)≤150 mg/kg total Zn in feedYes: Requires nano-specific bioavailability assessment (since 2021)
FDA (US)GRAS status (21 CFR §573.920)No: Applies to ZnO compounds generically
GB Standard (China)120–250 mg/kg total ZnPartial: Recommends NP accumulation monitoring
OECDTG 305:
Bioaccumulation test
Under revision: 2023 draft adds agreement on nanomaterials
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, J.; Xiong, D.; Long, M. Zinc Oxide Nanoparticles as Next-Generation Feed Additives: Bridging Antimicrobial Efficacy, Growth Promotion, and Sustainable Strategies in Animal Nutrition. Nanomaterials 2025, 15, 1030. https://doi.org/10.3390/nano15131030

AMA Style

Yang J, Xiong D, Long M. Zinc Oxide Nanoparticles as Next-Generation Feed Additives: Bridging Antimicrobial Efficacy, Growth Promotion, and Sustainable Strategies in Animal Nutrition. Nanomaterials. 2025; 15(13):1030. https://doi.org/10.3390/nano15131030

Chicago/Turabian Style

Yang, Jiayi, Dongwei Xiong, and Miao Long. 2025. "Zinc Oxide Nanoparticles as Next-Generation Feed Additives: Bridging Antimicrobial Efficacy, Growth Promotion, and Sustainable Strategies in Animal Nutrition" Nanomaterials 15, no. 13: 1030. https://doi.org/10.3390/nano15131030

APA Style

Yang, J., Xiong, D., & Long, M. (2025). Zinc Oxide Nanoparticles as Next-Generation Feed Additives: Bridging Antimicrobial Efficacy, Growth Promotion, and Sustainable Strategies in Animal Nutrition. Nanomaterials, 15(13), 1030. https://doi.org/10.3390/nano15131030

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

Article Metrics

Back to TopTop