Next Article in Journal
Current Advancements and Future Perspectives of Immunotherapy in Breast Cancer Treatment
Next Article in Special Issue
Anti-Inflammatory Efficacy of Resveratrol-Enriched Rice Callus Extract on Lipopolysaccharide-Stimulated RAW264.7 Macrophages
Previous Article in Journal
Immunopathogenesis of Nipah Virus Infection and Associated Immune Responses
Previous Article in Special Issue
Anti-Inflammatory and Anti-Adipogenesis Effects of Alchemilla vulgaris L., Salvia officinalis L., and Vitis vinifera L. in THP-1-Derived Macrophages and 3T3-L1 Cell Line
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Medicinal Plant Enriched Metal Nanoparticles and Nanoemulsion for Inflammation Treatment: A Narrative Review on Current Status and Future Perspective

Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Immuno 2023, 3(2), 182-194; https://doi.org/10.3390/immuno3020012
Submission received: 29 March 2023 / Revised: 20 April 2023 / Accepted: 24 April 2023 / Published: 29 April 2023

Abstract

:
Inflammation is considered a natural reaction of the immune system that can be caused by several factors such as pathogens, chemical substances, and damaged cells. Since the classical era, therapeutic substances have been made from medicinal plants. According to recent studies, nanotechnology provides a fresh approach to maintaining the standard quality, distribution, and bioactivity of therapeutic compounds. This review emphasizes the anti-inflammatory effects of green, synthetic, plant-based nanoparticles and nanoemulsions. A reduction of the dosage of anti-inflammatory medications and an improved therapeutic impact is highly desirable with an efficient drug delivery method. Along with the discussion of nanotechnology of medicinal plant-based anti-inflammatory effects, this review also offers a perspective view of the use of nanoparticles and nanoemulsions in inflammatory diseases in the future.

1. Introduction

Nanotechnology is focused on the synthesis of materials and small-scale (1–100 nm) manipulation of plant-based, chemical, and microbial materials and diverse nanoparticle types have shown novel features that may open up new therapeutic possibilities [1]. Due to their small size and favorable interactions with the cellular membrane, receptors, organic molecules, and amino acids, nanoparticles are widely used in biomedicine [2]. Therapeutic plants enriched with active metabolites are shown to be an affordable approach to eco-friendly metal nanoparticle manufacturing and can decrease the metal ion to Cu3+ and Pt2+, as poor solubility and less absorption in the human body can be resolved by encapsulating the active form of the medicinal plant as a nanocapsule by a different method for various biomedical applications such as inflammation [3].
Among various disorders, the inflammatory reaction is a key pathogenic component. While defending the host from inflammatory processes, the innate immune reaction also activates the innate inflammatory response system. Inflammatory and immunological responses can be regulated with the aid of anti-inflammatory medications [4]. The use of nanoparticles and nanoemulsions to target inflammation through the identification of molecules highly expressed on the surface of inflammatory cytokines or endothelial cells through increased vasculature permeability, or even through biomimicry, offers a promising treatment for inflammatory diseases [5]. This review offers a comprehensive insight into medicinal plant-based nanotechnology and outlines the various formation (Figure 1) and sizes of nanoparticles with their anti-inflammatory capabilities.

2. Synthesis of Medicinal Plant-Based Metal Nanoparticles and Nanoemulsion

The need for metal nanoparticle synthesis grows along with the number of applications, which are expanding daily. A different way of creating biocompatible NPs known as “green synthesis” has emerged in response to the difficulties in the traditional chemical synthesis process [6]. Utilizing plant extracts is a relatively straightforward approach to making nanoparticles, and it is one of the accessible green methods of synthesis for metal/metal oxide nanoparticles. Water is consistently regarded as the best and most appropriate solvent solution for synthesis operations to dissolve plant extracts [7]. Plant extracts are regarded as a superior and safe source for the creation of metal nanoparticles. Various phytochemicals present in medicinal plants are responsible for the reduction of metal ions present in metal nanoparticles. Metal ions change into metal atoms and change color and form nanoparticles characterized by several techniques (TEM, SEM, FTIR, and UV-Vis spectroscopy) [8].
Nanoemulsions are incredibly flexible systems when it comes to encapsulating components that dissolve in the disperse phase. Most typically, hydrophobic molecules from herbal plant extracts are necessary to keep the tiny drops of a dispersed phase from floating around in a continuous phase. In three different types of nanoemulsion, the o/w type nanoemulsion consists of the water phase and oil phase. Then, surfactants are used for both hydrophilic and hydrophobic parts which stabilize the droplet emulsion and prevent phase separation [9,10]. Due to the low energy requirements, easy processing, low cost, and small droplets, the ultrasonic homogenizers method is commonly used for the synthesis of nanoemulsion. Other methods include high-pressure homogenization, micro fluidization, and the inversion technique [11].

3. Anti-Inflammatory Properties of Metal Nanoparticles and Nanoemulsion

Throughout the past few decades, nanomedicine has become recognized as a promising anti-inflammatory agent. One of the most popular methods for creating nanoparticles is plant-mediated “green” synthesis, which typically requires a neutral pH and occurs at room temperature [12]. Macrophages ingest the debris from cells and tissue during inflammation through a process known as phagocytosis and support inflammation by producing activation signals such as LPS, interferon, interleukin, etc., that activate macrophages. When there is inflammation, neutrophils migrate to the area and produce pro-inflammatory mediators that draw macrophages to the area [13]. Nanomedicine has a stronger ability to penetrate epithelial cells and inflammatory cells, which improves the treatment’s efficiency and endurance [14]. In this review, we will only focus on medicinal plant-based metal nanoparticles and nanoemulsion mechanisms (Figure 2) and a brief discussion about that.
Reactive oxygen species (ROS) are by-products of the electron transport chain (ETC) and are created by NADPH oxidases found in phagocytes. Cellular lipids and proteins are oxidized by ROS, which damages DNA, encouraging endothelial dysfunction. Gold nanoparticles reduce ROS production and are an anti-inflammatory substance [15]. AuNPs decrease the release of IL17 and TNF-α which is stimulated by LPS from splenocytes. The MAPK pathway activates toll-like receptors from monocytes and some kinase which alter gene expression. The PI3K pathway inhibits LPS-induced TNF-α and activates NF-κB. Here, AuNPs downregulate cytokine production by modulating the MAPK and PI3K signaling pathways [16,17]. Silver nanoparticles inhibit vascular endothelial growth factor which stimulates T helper cell-induced inflammation and secretes pro-inflammatory cytokines. AgNPs also inhibit the production of COX2 gene expression by decreasing the secretion of IL-12 and TNF-α [18,19].
Similar to a silver nanoparticle, Zn nanoparticles inhibit both NF-κB and the caspase-1 enzyme in stimulated mast cells [20]. ZnNPs reduce the cytosolic degradation of IκBα, a cellular protein that suppresses NF-κB transcription, as well as NF-κB nuclear translocation which is triggered by LPS. In other ways, ZnNPs prevent mast cell proliferation and decrease LPS-induced COX-2 activation along with suppressing NO generation by IFN- yin LPS-activated macrophages [21,22]. Copper nanoparticles prevent inflammation by efficiently assisting in membrane stability by limiting the release of RBC lysosomal enzymes to specific amounts in most of the studies [23].
Nanoemulsions (NE) are a promising method for improving the oral bioavailability of drugs with poor solubility. [24]. Enhanced oral bioavailability is responsible for increasing the solubility of plant-extract-based NE into the gut and a reduction of the droplet size in the formulation. Nanoemulsions also prevents inflammation by blocking NF-κB and MCP-1 to prevent macrophage migration. Likewise, the study revealed that a nanoemulsion is also capable of downregulating the MAPK and NF-κB signaling cascade depending on the dosage at a molecular level [25,26] (Figure 3).

4. Medicinal Plant-Based Metal Nanoparticle

Currently, there has been a considerable increase in the production of NPs from therapeutic plants, which are crucial for the synthesis of nano drugs. Moreover, medicinal plants are considered a dependable and essential source of natural bioactive chemicals. The use of NPs has significantly improved nanoparticles and nanoemulsion, particularly in terms of lowering the dose frequency, enhancing drug solubility, and lengthening the half-life of some medications, leading to significant advances in targeted drug delivery [27]. Medicinal plants are extracts rich in proteins, carbohydrates, terpenoids, bioactive alkaloids, and phenolic acids that can reduce the metallic ions and stabilize them [28,29].

4.1. Therapeutic Gold and Ag Nanoparticles in Inflammation

Gold nanoparticles (AuNPs) have been utilized effectively for drug delivery and have well-established bioactivities. Gold nanoparticles (GNPs or AuNPs) appear to be the most efficient nanoparticles studied in experimental research, with the least systemic toxicity. AuNPs reduce NO generation and iNOS expression in RAW264.7 cells activated with LPS by preventing NF-kB and STAT1 activation [30]. Numerous research studies have revealed that Panax ginseng Meyer may have minimal to no negative side effects and have potentially helpful impacts on a range of illnesses and human health. Natural materials used in biologically generated AuNPs exhibit a variety of benefits that present different phytochemicals found in plants [31,32]. Synthesized Panax ginseng leaf extract-mediated gold nanoparticles (10–20 nm) inhibit the expression of NF-kB in LPS-induced RAW 264 [33] cells through the blocking of the MAPK pathway. Another medicinal plant named Suaeda japonica is a halophytic herb originating from Korea and Japan, and has been used to synthesize gold nanoparticles which are 20–30 nm and spherical. S. japonica successfully revealed its anti-inflammatory potential by reducing the release of pro-inflammatory cytokines and suppressing the expression of the genes iNOS, COX-2, and TNF-a [34,35].
As a first line of defense, the skin plays a crucial role in the immune system against different environmental stimuli and acts as tissue homeostasis. Moreover, an aberrant skin immune response potentially leads to inflammatory skin diseases [36,37]. Medicinal plant Rosa Rogusa-mediated gold nanoparticles exhibit significant anti-inflammatory effects in keratinocytes. The study revealed a novel approach to synthesizing RRAuNPs which were only 38.2 mm in size and significantly downregulated the gene expression of RANTES, TARC, CTACK, IL-6, and IL-8 of TNF-α/IFN-γ-induced HaCaT cells [38].
Natural killer cells possess the ability to both stimulate and limit adaptive immune systems that could otherwise result in excessive inflammation or even autoimmunity [39]. Innate and adaptive immune cell recruitment in chronic inflammation produces the synthesis of significant amounts of pro-inflammatory modulators. Elbagory et al. demonstrated that gold nanoparticles based on the medicinal plant Hypoxis Hemerocallidea suppressed pro-inflammatory cytokines production in THP1 cells and prevented chronic inflammation involving NK cells [40,41].
Nearly every component of an organism’s immunological reaction involves macrophages. Localized macrophages control inflammation by serving as sentinels and reacting to internal and external physiological changes [42]. As autophagy is considered an important component of innate immunity, inflammatory and viral disorders can be made more likely by autophagy dysfunction [43,44]. Damaged mitochondria accumulated as a result of an impaired autophagic flux could produce too much ROS and lead to cell death [45]. Recent research reported medicinal plant Hibiscus Syriacus L-mediated gold nanoparticles acted as an inducer of autophagy for LPS-induced macrophage inflammation. HCE-NPs reduced the overproduction of pro-inflammatory mediators in RAW264.7 cells after LPS-induced inflammation via an autophagy-dependent approach. By decreasing ROS generation and restoring the levels of ATP, NPs prevent accelerated inflammation [46]. We have summarized some of the interesting research works on the anti-inflammatory effects of current gold and silver nanoparticles in Table 1.
A growing number of sectors, including those in medicine, food, health care, consumer goods, and industrial uses, are using silver nanoparticles (AgNPs) based on distinctive physical and chemical characteristics [52]. AgNPs are produced using multiple strategies, such as chemical and physical ones; however, they all involve the employment of dangerous substances and high temperatures [53,54]. Researchers’ curiosity has been piqued by the special properties of plant-based silver nanoparticles, such as non-toxic efficiency [55,56]. Research has shown that silver nanoparticles synthesized from Azadirachta indica kernel aqueous extract have a significant anti-inflammatory effect in vitro. A variety of biological activities make Azadirachta indica one of the most useful therapeutic herbs. Spherical-shaped 19–22 nm sized green synthesized AgNPs control protein degradation dose-dependently which prevents arthritic inflammation [47]. Another traditional plant named Cotyledon orbiculate from South Africa is very notable to treat skin inflammation. Synthesized stable silver nanoparticles from Cotyledon orbiculate exert immunomodulatory effects in the THP-1 macrophage by a reduction of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β [48].
Asparagus racemosus has been employed in conventional treatments for several immune-related illnesses [57]. The study discovered that medicinal A. racemosus biosynthesized mixed Ag–Au bimetallic alloy nanoparticles had significant effects on NK92 cells than only synthesized gold or silver nanoparticle aqueous extract, considered as an anti-inflammatory response. In that study, IFN-y release was dramatically decreased, and the cytokine response was impacted by the Ag–Au bimetallic alloy nanoparticles in NK92 compared to individual nanoparticles or extract [58].
Stein Leventhal syndrome, also referred to as PCOS (polycystic ovary syndrome), is an inflammatory condition that causes metabolic imbalance, ovarian dysfunction, and infertility in women. Specific mediators of inflammation (IL-6, IL-18, and TNF-α) are elevated in PCOS women, which points to a change in their immune system [59]. A Cinnamomum zeylanicum-synthesized silver nanoparticle study revealed a reduction of an inflammatory biomarker in PCOS in female rats [49].

4.2. Medicinal Plant-Based Zinc and Copper Nanoparticles and Their Impact on Inflammation

ZnO NPs have been produced using a range of plants, microbes, algae, fungi, and other biological materials such as starch and eggs. The plant technique provides many benefits over the microbe strategy since it does not require isolation, culture development, or maintenance [60]. The primary mechanism of action involves a phytochemical present in natural extracts oxidizing and reducing zinc ions. The strategy of green synthesis also demonstrates an increased catalytic process and minimizes exposition to the use of dangerous and pricey chemicals, which can aid in reducing their toxicity to the environment [61].
Recent work investigated the anti-inflammatory effects of the medicinal plant Terminalia ferdinandiana (Kakadu plum). (ZnO) nanoparticles (NPs) were flower shaped with an average size of 21.99 nm. Nitric oxide (NO), a main mediator during inflammatory reactions, is produced by macrophages. It is created from the amino acid arginine by the enzyme inducible nitric oxide synthase (iNOS). ZnO nanoparticles formed from the Kakadu plum successfully suppressed the release of nitric oxide in LPS-triggered raw 264.7 cells [62,63]. According to Vijaykumar et al., Anoectochilus elatus leaf extracts were used to develop zinc nanoparticles and investigate their anti-inflammatory properties. An in vitro assay by ZnNPs revealed the inhibition of protein (albumin) denaturation and protease activity compared to the standard drug aspirin in the microbiome [64]. Another popular medicinal plant, Zingiber officinale, was used to develop ZnO nanoparticles. This bioinspired nanoparticle is used to check inflammatory activity through an enzyme assay. Usually, prostaglandins cause swelling, discomfort, fever, and redness at the injection site when there is inflammation. Ginger-capped ZnNPs inhibit COX1 and COX2 enzymes which are responsible for PG production and act as a potential anti-inflammatory response [65,66]. Dhivyadharshin et al. reported another zinc oxide nanoparticle formation from the Unani plant Adhatoda vasica, which showed significant anti-inflammatory efficacy by protein degradation [67]. We have illustrated some of the research activities on the current copper and zinc nanoparticles in Table 2.
Besides zinc, copper is commonly used to synthesize nanoparticles due to its distinct physical and molecular characteristics such as a large surface area, strength, flexibility, and rigidity [73]. Many studies have reported the anti-inflammatory efficacy of copper nanoparticles. Among them, traditional herbal plant-based copper nanoparticles such as Myrtus Communis leaves extract-formed CuNPs attained maximum level albumin inhibition compared to the standard drug aspirin [70]. Similarly, Mucuna pruriens seed extract-mediated CuNPs can inhibit protein denaturation dose-dependently and act as a potential drug for inflammation [71]. Some research with CuNPs focused on the anti-inflammatory activity via the membrane stabilization method. Copper nanoparticles using Cissus quadrangularis and Mussaenda frondosa L were used on red blood cells where these nanoparticles efficiently assisted in membrane stability by limiting the release of RBC lysosomal enzymes to specific amounts [74,75].

5. Plant-Based Nanoemulsion for Inflammation

Nanoemulsions are now appealing nanocarriers among other nanoparticles due to their capacity to increase drug delivery via bio-membranes, lengthen the half-life (t1/2) in the body, and encapsulate drugs with a high lipophilic capability. Several studies support the oil in water (O/W) type of nanoemulsion as an effective carrier to solubilize the hydrophobic chemical in their oily cores among the three varieties of nanoemulsions (o/w, w/o, and w/o/w) [76]. Recently, nanoemulsions have become widely used to safeguard plant active ingredients from harsh environments, improve medicine solubility and stability, and heighten drug efficacy [77]. When compared to traditional preparation, the encapsulation of herbal plant components is an effective method for delivering NE and can include a greater amount of medicines [78]. The study revealed that Panax ginseng, which is known as the king of medicinal plants, successfully formed NE with sea buckwheat fruit oil. That study also demonstrated the inhibition of pro-inflammatory mediators in vitro compared to Panax ginseng alone [79,80]. Previously, an in vivo study in a mouse ear model with curcumin NE (formed with MCT) from the Curcuma longa medicinal plant focused on the inhibition effects of edema as an anti-inflammatory potential [81]. Table 3 has summarized the recent work on medicinal plant-based nanoemulsions and their anti-inflammatory activities.
By exhibiting the immunomodulatory activity and preventing the generation of nitric oxide, a pro-inflammatory agent, NE has additionally shown the ability to enhance the anti-inflammatory effects of medicinal plant-based essential oils. A Rosmarinus officinalis L-derived essential oil was used to make an O/W NE and anti-inflammatory activity was demonstrated in zebrafish [82]. Moreover, an advanced approach has been made in recent research where a nanoemulsion was made with nanoparticles and exhibited an anti-inflammatory capacity. A mountain ginseng-based gold nanoparticle was used to prepare NE with another medicinal plant constituting silydianin and investigated potent anti-inflammatory activity in Raw 264.7 cells through MAPK downregulation and NF-κB signaling pathways [83]. The phytocompound of the flower extract of the herbal plant Woodfordia fruticose K synthesized a nanoemulsion and revealed protein denaturation which assured further tissue inflammation and cell stabilization potential in a human red blood cell membrane [84].

6. Current Status, Limitation, and Future Perspective of Metal-Based Nanoparticles and Nanoemulsion for Inflammation

In the past two decades, considerable study has been conducted on the biosynthesis of metal nanoparticles utilizing plant derivatives. Plant metabolites encourage the environmentally beneficial creation of metallic nanoparticles. The current study provides a noteworthy and succinct report on the anti-inflammatory strategies used by different medicinal plant-based nanoparticles, including silver, gold, zinc oxide, and nanoemulsion. It is unnecessary to use hazardous chemicals as reducing and capping agents when synthesizing nanoparticles from plant sources. This process is also more economical and beneficial to the environment. Nanomedicine has already been proven to have a stronger ability to penetrate epithelial cells and inflammatory cells, which improves the treatment’s efficiency and endurance by choosing the probable target site. Moreover, green-synthesized nanoparticles had a more potent impact on inflammation than conventionally synthesized nanoparticles. Different interactions between nanomedicines and inflammatory efficacy in cells, common mechanisms involved in the anti-inflammatory activity, and the synthesis of herbal nanoparticles and nanoemulsions has been briefly discussed in this research.
Although used for various biomedical applications, especially for cancer, inflammation, and radiotherapy, metal nanoparticles’ immunotoxicity has been inconsistent and even contradictory. Immune toxicity is defined as the harmful effect on the immune system as the chronic inflammation of immunosuppression and autoimmune diseases [87,88]. Despite the potential biomedical application of metal NPs, biohazards and toxicities remain unclear.
Among the metal Nps, ZnO Nps are used in biomedical applications but due to their unique physicochemical properties, they could easily access several immune tissues and cells by inhalation and skin uptake. Further, oral administration could cause severe damage to several organs such as the kidney, heart, and liver. Size and charge are crucial factors for enhancing toxicity. The negative charge is less toxic in vitro Raw264.7 cells than positive ZnO NPs [89,90,91,92]. The different sized and charged ZnO and CuO NPs would cause in vitro and in vivo immunotoxicity, of which their nature is immunosuppression. To solve the issue further, there is a need to find the surface properties that greatly affect the toxicity and interaction of the organ and cell before evaluating the toxicity (geno toxicity/cytotoxicity/immunotoxicity) of the nanomaterial parameters such as morphology, size, chemical composition, surface properties (chemistry, charge) mechanism, or pathways that are involved in toxicity. On the other hand, nanoemulsions also have toxicity during oral administration due to their ability to change the biological fate of bioactive components within the gastrointestinal tract. Some of these lipophilic components are typically digested within the human gastrointestinal (GI) tract (e.g., triacylglycerols), some normally pass through the GI tract without being absorbed (e.g., mineral oils and fat substitutes), while some are absorbed without being digested [93].
As per the future strategy, more research is needed to find feasible molecules to synthesize metal (gold, silver, zinc, and copper) for commercialization. In addition, relevant research should be conducted in vivo on green metal nanoparticles and used as a safe medication in inflammatory disorders and progressive chronic inflammation.
Nanoemulsions have drawn great interest over the past few decades for a variety of applications. Many herbal plant compounds have been used to develop emulsions in nano size and improve bioavailability with anti-inflammatory potential. Mostly, nanoemulsions have been developed to encapsulate the hydrophobic green components of plants and deliver them to the target site efficiently. This study briefly demonstrated medicinal plant-based nanoemulsions, their mechanism, and their efficacy in inflammation. This study also noticed some research on nanoparticle-encapsulated nanoemulsion formation preventing inflammatory disorder.
In the preparation of nanoemulsions using some organic solvents, such as acetone, hexane, or ethyl acetate, those solvents used are removed by evaporation during the preparation of the nanoemulsion, but some residual solvents may remain in the final product [94]. It is therefore important to be aware of the potential toxic effects associated with any residual organic solvents if nanoemulsions are produced using this approach.
In the future world, nanoemulsions could be used as a vaccine delivery as well as immunotherapy for a genetic disorder. So, further research is needed to evaluate the inflammatory efficacy of potential hydrophobic medicinal plant parts with bio formation by nanoemulsions in an animal and clinical trial. It is urgently necessary to make progress in this area since it will be necessary to build sustainable nanotechnology for vaccines and other biological applications. Above all, it is urgently crucial to make progress in this area since it will be necessary to build sustainable nanotechnology not only to treat inflammation but also for developing a vaccine, immune-enhancing drug, and chemotherapy.

Author Contributions

A.M.P. Conceptualization, writing—original draft preparation, figure modeling, E.J.R.: writing—draft and editing, Y.J.K.: supervision, review, and editing, D.-C.Y.: review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by KDBIO Corp. and also supported by the fund “National Research Foundation of Korea (NRF) funded by the Ministry of Education (2023R1A2C1007606)”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chaloupka, K.; Malam, Y.; Seifalian, A.M. Nanosilver as a new generation of nanoproduct in biomedical applications. Trends Biotechnol. 2010, 28, 580–588. [Google Scholar] [CrossRef] [PubMed]
  2. Yu, Z.; Li, Q.; Wang, J.; Yu, Y.; Wang, Y.; Zhou, Q.; Li, P. Reactive oxygen species-related nanoparticle toxicity in the biomedical field. Nanoscale Res. Lett. 2020, 15, 1–14. [Google Scholar] [CrossRef]
  3. Paramanya, A.; Sharma, S.; Bagdat, R.B.; Ali, A. Recent practices of medicinal and aromatic plants in nanotechnology. In Nanomaterials for Agriculture and Forestry Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 435–467. [Google Scholar]
  4. Ferrero-Miliani, L.; Nielsen, O.H.; Andersen, P.S.; Girardin, S.E. Chronic inflammation: Importance of NOD2 and NALP3 in interleukin-1beta generation. Clin. Exp. Immunol. 2007, 147, 227–235. [Google Scholar] [CrossRef]
  5. Brusini, R.; Varna, M.; Couvreur, P. Advanced nanomedicines for the treatment of inflammatory diseases. Adv. Drug Deliv. Rev. 2020, 157, 161–178. [Google Scholar] [CrossRef]
  6. Soltys, L.; Olkhovyy, O.; Tatarchuk, T.; Naushad, M. Green Synthesis of Metal and Metal Oxide Nanoparticles: Principles of Green Chemistry and Raw Materials. Magnetochemistry 2021, 7, 145. [Google Scholar] [CrossRef]
  7. Shanker, U.; Jassal, V.; Rani, M.; Kaith, B.S. Towards green synthesis of nano-particles: From bio-assisted sources to benign solvents. A review. Int. J. Environ. Anal. Chem. 2016, 96, 801–835. [Google Scholar]
  8. Singh, J.; Dutta, T.; Kim, K.H.; Rawat, M.; Samddar, P.; Kumar, P. ‘Green’ synthesis of metals and their oxide nanoparticles: Applications for environmental remediation. J. Nanobiotechnol. 2018, 16, 1–24. [Google Scholar] [CrossRef]
  9. Javadi, S.; Kazemi, N.M.; Halabian, R. Preparation of O/W nano-emulsion containing nettle and fenugreek extract and cumin essential oil for evaluating antidiabetic properties. AAPS Open 2021, 7, 13. [Google Scholar] [CrossRef]
  10. Elzayat, A.; Adam-Cervera, I.; Álvarez-Bermúdez, O.; Muñoz-Espí, R. Nanoemulsions for synthesis of biomedical nanocarriers. Colloids Surf. B Biointerfaces 2021, 203, 111764. [Google Scholar] [CrossRef] [PubMed]
  11. Gharibzahedi, S.M.; Jafari, S.M. Fabrication of nanoemulsions by ultrasonication. In Nanoemulsions; Academic Press: Cambridge, MA, USA, 2018; pp. 233–285. [Google Scholar]
  12. Velusamy, P.; Kumar, G.V.; Jeyanthi, V.; Das, J.; Pachaiappan, R. Bio-inspired green nanoparticles: Synthesis, mechanism, and antibacterial application. Toxicol. Res. 2016, 32, 95–102. [Google Scholar] [CrossRef]
  13. Fujiwara, N.; Kobayashi, K. Macrophages in Inflammation. Curr. Drug Targets Inflamm. Allergy 2005, 3, 281–286. [Google Scholar] [CrossRef]
  14. Viscido, A.; Capannolo, A.; Latella, G.; Caprilli, R.; Frieri, G. Nanotechnology in the treatment of inflammatory bowel diseases. J. Crohns Colitis 2014, 8, 903–918. [Google Scholar] [CrossRef]
  15. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef]
  16. Sumbayev, V.V.; Yasinska, I.M.; Garcia, C.P.; Gilliland, D.; Lall, G.S.; Gibbs, B.F.; Bonsall, D.R.; Varani, L.; Rossi, F.; Calzolai, L. Gold nanoparticles downregulate interleukin-1β-induced pro-inflammatory responses. Small 2013, 9, 472–477. [Google Scholar] [CrossRef]
  17. de Carvalho, T.G.; Garcia, V.B.; de Araújo, A.A.; da Silva Gasparotto, L.H.; Silva, H.; Guerra, G.C.B.; de Castro Miguel, E.; de Carvalho Leitão, R.F.; da Silva Costa, D.V.; Cruz, L.J.; et al. Spherical neutral gold nanoparticles improve anti-inflammatory response, oxidative stress and fibrosis in alcohol-methamphetamine-induced liver injury in rats. Int. J. Pharm. 2018, 548, 1–14. [Google Scholar] [CrossRef]
  18. Lee, C.G.; Link, H.; Baluk, P.; Homer, R.J.; Chapoval, S.; Bhandari, V.; Kang, M.J.; Cohn, L.; Kim, Y.K.; McDonald, D.M.; et al. Vascular endothelial growth factor (VEGF) induces remodeling and enhances T(H)2-mediated sensitization and inflammation in the lung. Nat. Med. 2004, 10, 1095–1103. [Google Scholar] [CrossRef] [PubMed]
  19. Franková, J.; Pivodová, V.; Vágnerová, H.; Juráňová, J.; Ulrichová, J. Effects of silver nanoparticles on primary cell cultures of fibroblasts and keratinocytes in a wound healing model. J. Appl. Biomater. Funct. Mater. 2016, 14, 137–142. [Google Scholar] [CrossRef]
  20. Kim, M.H.; Jeong, H.J. Zinc oxide nanoparticles suppress LPS-Induced NF- κB activation by inducing A20, a negative regulator of NF- κ B, in RAW 264.7 macrophages. J. Nanosci. Nanotechnol. 2015, 9, 6509–6515. [Google Scholar] [CrossRef]
  21. 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]
  22. Tripathi, P.; Tripathi, P.; Kashyap, L.; Singh, V. The role of nitric oxide in inflammatory reactions. FEMS Immunol. Med. Microbiol. 2007, 51, 443–452. [Google Scholar] [CrossRef] [PubMed]
  23. Govindappa, M.; Hemashekhar, B.; Arthikala, M.K.; Rai, V.R.; Ramachandra, Y.L. Characterization, antibacterial, antioxidant, antidiabetic, anti-inflammatory and antityrosinase activity of green synthesized silver nanoparticles using Calophyllum tomentosum leaves extract. Results Phys. 2018, 9, 400–408. [Google Scholar] [CrossRef]
  24. Yen, C.C.; Chen, Y.C.; Wu, M.T.; Wang, C.C.; Wu, Y.T. Nanoemulsion as a strategy for improving the oral bioavailability and anti-inflammatory activity of andrographolide. Int. J. Nanomed. 2018, 13, 669. [Google Scholar] [CrossRef]
  25. Shakibaei, M.; John, T.; Schulze-Tanzil, G.; Lehmann, I.; Mobasheri, A. Suppression of NF-kappaB activation by curcumin leads to inhibition of expression of cyclo-oxygenase-2 and matrix metalloproteinase-9 in human articular chondrocytes: Implications for the treatment of osteoarthritis. Biochem. Pharmacol. 2007, 73, 1434–1445. [Google Scholar] [CrossRef] [PubMed]
  26. Bachstetter, A.D.; Van Eldik, L.J. The p38 MAP kinase family as regulators of proinflammatory cytokine production in degenerative diseases of the CNS. Aging Dis. 2010, 1, 199–211. [Google Scholar]
  27. Xulu, J.H.; Ndongwe, T.; Ezealisiji, K.M.; Tembu, V.J.; Mncwangi, N.P.; Witika, B.A.; Siwe-Noundou, X. The Use of Medicinal Plant-Derived Metallic Nanoparticles in Theranostics. Pharmaceutics 2022, 14, 2437. [Google Scholar] [CrossRef]
  28. Shah, M.; Fawcett, D.; Sharma, S.; Tripathy, S.K.; Poinern, G.E.J. Green Synthesis of Metallic Nanoparticles via Biological Entities. Materials 2015, 8, 7278–7308. [Google Scholar] [CrossRef]
  29. Bharadwaj, K.K.; Rabha, B.; Pati, S.; Sarkar, T.; Choudhury, B.K.; Barman, A.; Bhattacharjya, D.; Srivastava, A.; Baishya, D.; Edinur, H.A.; et al. Green Synthesis of Gold Nanoparticles Using Plant Extracts as Beneficial Prospect for Cancer Theranostics. Molecules 2021, 26, 6389. [Google Scholar] [CrossRef] [PubMed]
  30. Ma, J.S.; Kim, W.J.; Kim, J.J.; Kim, T.J.; Ye, S.K.; Song, M.D.; Kang, H.; Kim, D.W.; Moon, W.K.; Lee, K.H. Gold nanoparticles attenuate LPS-induced NO production through the inhibition of NF-κB and IFN-β/STAT1 pathways in RAW264.7 cells. Nitric Oxide 2010, 23, 214–219. [Google Scholar] [CrossRef]
  31. Chandran, S.P.; Chaudhary, M.; Pasricha, R.; Ahmad, A.; Sastry, M. Synthesis of gold nanotriangles and silver nanoparticles using Aloevera plant extract. Biotechnol. Prog. 2006, 22, 577–583. [Google Scholar] [CrossRef]
  32. Hwang, S.J.; Jun, S.H.; Park, Y.; Cha, S.-H.; Yoon, M.; Cho, S.; Lee, H.-J.; Park, Y. Green synthesis of gold nanoparticles using chlorogenic acid and their enhanced performance for inflammation. Nanomedicine 2015, 11, 1677–1688. [Google Scholar] [CrossRef] [PubMed]
  33. Ahn, S.; Singh, P.; Castro-Aceituno, V.; Yesmin Simu, S.; Kim, Y.J.; Mathiyalagan, R.; Yang, D.C. Gold nanoparticles synthesized using Panax ginseng leaves suppress inflammatory-mediators production via blockade of NF-κB activation in macrophages. Artif. Cells Nanomed. Biotechnol. 2017, 45, 270–276. [Google Scholar] [CrossRef]
  34. Cho, J.-Y.; Yang, X.; Park, K.-H.; Park, H.J.; Park, S.-Y.; Moon, J.-H.; Ham, K.-S. Isolation and identification of antioxidative compounds and their activities from Suaeda japonica. Food Sci. Biotechnol. 2013, 22, 1547–1557. [Google Scholar] [CrossRef]
  35. Kwak, G.Y.; Han, Y.; Baik, S.; Kong, B.M.; Yang, D.C.; Kang, S.C.; Sukweenadhi, J. Green-Synthesized Gold Nanoparticles by the Suaeda japonica Leaf Extract and Screening of Anti-Inflammatory Activities on RAW 267.4 Macrophages. Coatings 2022, 12, 460. [Google Scholar] [CrossRef]
  36. Nguyen, A.V.; Soulika, A.M. The dynamics of the skin’s immune system. Int. J. Mol. Sci. 2019, 20, 1811. [Google Scholar] [CrossRef] [PubMed]
  37. Juráňová, J.; Franková, J.; Ulrichová, J. The role of keratinocytes in inflammation. J. Appl. Biomed. 2017, 15, 169–179. [Google Scholar] [CrossRef]
  38. Wang, R.; Moon, S.K.; Kim, W.J.; Dhandapani, S.; Kim, H.; Kim, Y.J. Biologically Synthesized Rosa rugosa-Based Gold Nanoparticles Suppress Skin Inflammatory Responses via MAPK and NF-κB Signaling Pathway in TNF-α/IFN-γ-Induced HaCaT Keratinocytes. ACS Omega 2022, 7, 35951–35960. [Google Scholar] [CrossRef]
  39. Zitti, B.; Bryceson, Y.T. Natural killer cells in inflammation and autoimmunity. Cytokine Growth Factor Rev. 2018, 42, 37–46. [Google Scholar] [CrossRef]
  40. Meirow, Y.; Baniyash, M. Immune biomarkers for chronic inflammation related complications in non-cancerous and cancerous diseases. Cancer Immunol. Immunother. 2017, 66, 1089–1101. [Google Scholar] [CrossRef] [PubMed]
  41. Elbagory, A.M.; Hussein, A.A.; Meyer, M. The in vitro immunomodulatory effects of gold nanoparticles synthesized from Hypoxis hemerocallidea aqueous extract and hypoxoside on macrophage and natural killer cells. Int. J. Nanomed. 2019, 14, 9007–9018. [Google Scholar] [CrossRef]
  42. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455. [Google Scholar] [CrossRef]
  43. Yu, L.; Chen, Y.; Tooze, S.A. Autophagy pathway: Cellular and molecular mechanisms. Autophagy 2018, 14, 207–215. [Google Scholar] [CrossRef]
  44. Zhao, T.; Zheng, T.; Yu, H.; Hu, B.H.; Hu, B.; Ma, P.; Yang, Y.; Yang, N.; Hu, J.; Cao, T.; et al. Autophagy impairment as a key feature for acetaminophen-induced ototoxicity. Cell Death Dis. 2021, 12, 3. [Google Scholar] [CrossRef] [PubMed]
  45. Ma, X.; Wu, Y.; Jin, S.; Tian, Y.; Zhang, X.; Zhao, Y.; Yu, L.; Liang, X.J. Gold nanoparticles induce autophagosome accumulation through size-dependent nanoparticle uptake and lysosome impairment. ACS Nano 2011, 5, 8629–8639. [Google Scholar] [CrossRef]
  46. Xu, X.Y.; Tran, T.H.M.; Perumalsamy, H.; Sanjeevram, D.; Kim, Y.J. Biosynthetic gold nanoparticles of Hibiscus syriacus L. callus potentiates anti-inflammation efficacy via an autophagy-dependent mechanism. Mater. Sci. Eng. C 2021, 124, 112035. [Google Scholar] [CrossRef] [PubMed]
  47. Chi, N.T.L.; Narayanan, M.; Chinnathambi, A.; Govindasamy, C.; Subramani, B.; Brindhadevi, K.; Pimpimon, T.; Pikulkaew, S. Fabrication, characterization, anti-inflammatory, and anti-diabetic activity of silver nanoparticles synthesized from Azadirachta indica kernel aqueous extract. Environ. Res. 2022, 208, 112684. [Google Scholar] [CrossRef]
  48. Tyavambiza, C.; Elbagory, A.M.; Madiehe, A.M.; Meyer, M.; Meyer, S. The antimicrobial and anti-inflammatory effects of silver nanoparticles synthesised from Cotyledon orbiculata aqueous extract. Nanomaterials 2021, 11, 1343. [Google Scholar] [CrossRef] [PubMed]
  49. Alwan, S.H.; Al-Saeed, M.H. Silver Nanoparticles Biofabricated from Cinnamomum zeylanicum Reduce IL-6, IL-18, and TNF-ɑ in Female Rats with Polycystic Ovarian Syndrome. Int. J. Fertil. Steril. 2023, 17, 80–84. [Google Scholar] [CrossRef]
  50. Chakravarty, A.; Ahmad, I.; Singh, P.; Sheikh, M.U.D.; Aalam, G.; Sagadevan, S.; Ikram, S. Green synthesis of silver nano-particles using fruits extracts of Syzygium cumini and their bioactivity. Chem. Phys. Lett. 2022, 795, 139493. [Google Scholar] [CrossRef]
  51. Kedi, P.B.E.; Meva, F.E.A.; Kotsedi, L.; Nguemfo, E.L.; Zangueu, C.B.; Ntoumba, A.A.; Mohamed, H.E.A.; Dongmo, A.B.; Maaza, M. Eco-friendly synthesis, characterization, in vitro and in vivo anti-inflammatory activity of silver nanoparticle-mediated Selaginella myosurus aqueous extract. Int. J. Nanomedicine. 2018, 13, 8537. [Google Scholar] [CrossRef]
  52. Zhang, X.F.; Liu, Z.G.; Shen, W.; Gurunathan, S. Silver nanoparticles: Synthesis, characterization, properties, applications, and therapeutic approaches. Int. J. Mol. Sci. 2016, 17, 1534. [Google Scholar] [CrossRef]
  53. Anusha, P.; Narayanan, M.; Natarajan, D.; Kandasamy, S.; Chinnathambi, A.; Alharbi, S.A.; Brindhadevi, K. Assessment of hexavalent chromium (VI) biosorption competence of indigenous Aspergillus tubingensis AF3 isolated from bauxite mine tailing. Chemosphere 2021, 282, 131055. [Google Scholar] [CrossRef]
  54. Hasan, N.; Ahmad, N.; Zohrameena, S.; Khalid, M.; Akhtar, J. Asparagus racemosus: For medicinal uses &pharmacological actions. Int. J. Adv. Res. 2016, 4, 259–267. [Google Scholar]
  55. Amina, M.; Al Musayeib, N.M.; Alarfaj, N.A.; El-Tohamy, M.F.; Al-Hamoud, G.A. Antibacterial and immunomodulatory potentials of biosynthesized Ag, Au, Ag-Au bimetallic alloy nanoparticles using the Asparagus racemosus root extract. Nanomaterials 2020, 10, 2453. [Google Scholar] [CrossRef] [PubMed]
  56. Fulghesu, A.M.; Sanna, F.; Uda, S.; Magnini, R.; Portoghese, E.; Batetta, B. IL-6 serum levels and production is related to an altered immune response in polycystic ovary syndrome girls with insulin resistance. Mediat. Inflamm. 2011, 2011, 389397. [Google Scholar] [CrossRef]
  57. Narayanan, M.; Gopi, A.; Natarajan, D.; Kandasamy, S.; Saravanan, M.; El Askary, A.; Elfasakhany, A.; Pugazhendhi, A. Hepato and nephroprotective activity of methanol extract of Hygrophila spinosa and its antibacterial potential against multidrug resistant Pandoraea sputorum. Environ. Res. 2021, 201, 111594. [Google Scholar] [CrossRef] [PubMed]
  58. Aziz, T.; Ullah, A.; Fan, H.; Ullah, R.; Haq, F.; Khan, F.U.; Iqbal, M.; Wei, J. Cellulose nanocrystals applications in health, medicine and catalysis. J. Polym. Environ. 2021, 29, 2062–2071. [Google Scholar] [CrossRef]
  59. Andarge, E.; Shonga, A.; Agize, M.; Tora, A. Utilization and conservation of medicinal plants and their associated indigenous knowledge (IK) in Dawuro Zone: An ethnobotanical approach. Int. J. Med. Plant Res. 2015, 4, 330–337. [Google Scholar]
  60. Fadiji, A.E.; Babalola, O.O. Metagenomics methods for the study of plant-associated microbial communities: A review. J. Microbiol. Methods 2020, 170, 105860. [Google Scholar] [CrossRef] [PubMed]
  61. Huang, Y.; Haw, C.Y.; Zheng, Z.; Kang, J.; Zheng, J.-C.; Wang, H.-Q. Biosynthesis of Zinc Oxide Nanomaterials from Plant Extracts and Future Green Prospects: A Topical Review. Adv. Sustain. Syst. 2021, 5, 200026. [Google Scholar] [CrossRef]
  62. Sharma, J.; Al-Omran, A.; Parvathy, S. Role of nitric oxide in inflammatory diseases. Inflammopharmacology 2007, 15, 252–259. [Google Scholar] [CrossRef]
  63. Ramadhania, Z.M.; Nahar, J.; Ahn, J.C.; Yang, D.U.; Kim, J.H.; Lee, D.W.; Kong, B.M.; Mathiyalagan, R.; Rupa, E.J.; Akter, R.; et al. Terminalia ferdinandiana (Kakadu Plum)-Mediated Bio-Synthesized ZnO Nanoparticles for Enhancement of Anti-Lung Cancer and Anti-Inflammatory Activities. Appl. Sci. 2022, 12, 3081. [Google Scholar] [CrossRef]
  64. Vijayakumar, N.; Bhuvaneshwari, V.K.; Ayyadurai, G.K.; Jayaprakash, R.; Gopinath, K.; Nicoletti, M.; Alarifi, S.; Govindarajan, M. Green synthesis of zinc oxide nanoparticles using Anoectochilus elatus, and their biomedical applications. Saudi J. Biol. Sci. 2022, 29, 2270–2279. [Google Scholar] [CrossRef] [PubMed]
  65. Scarano, D.; Bertarione, S.; Spoto, G.; Zecchina, A.; Areán, C.O. FTIR spectroscopy of hydrogen, carbon monoxide, and methane absorbed and co-adsorbed on zinc oxide. Thin Solid Film. 2001, 400, 50–55. [Google Scholar] [CrossRef]
  66. Al-Radadi, N.S.; Faisal, S.; Alotaibi, A.; Ullah, R.; Hussain, T.; Rizwan, M.; Zaman, N.; Iqbal, M.; Iqbal, A.; Ali, Z. Zingiber officinale driven bioproduction of ZnO nanoparticles and their anti-inflammatory, anti-diabetic, anti-Alzheimer, anti-oxidant, and anti-microbial applications. Inorg. Chem. Commun. 2022, 140, 109274. [Google Scholar] [CrossRef]
  67. Dhivyadharshini, J. Evaluation of anti-inflammatory and antioxidant activity of Adhatoda vasica zinc nanoparticles. Nveo-Nat. Vola. Essen. Oils. 2021, 8, 5950–5964. [Google Scholar]
  68. 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 2015, 146, 10–17. [Google Scholar] [CrossRef]
  69. Agarwal, H.; Shanmugam, V.K. 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. [Google Scholar] [CrossRef]
  70. Al-Jubouri, A.K.; Al-Saadi, N.H.; Kadhim, M.A. Anti-Inflammatory and Anti-Bacterial Activity of Copper Nanoparticles Synthesized from Myrtus Communis Leaves Extract. Iraqi J. Agric. Sci. 2022, 53, 698–711. [Google Scholar] [CrossRef]
  71. Anushya, P.; V Geetha, R.; Rajesh Kumar, S. Evaluation of Anti Inflammatory and Cytotoxic Effect of Copper Nanoparticles Synthesized Using Seed Extract of Mucuna pruriens. J. Pharm. Res. Int. 2021, 33, 816–824. [Google Scholar] [CrossRef]
  72. Liu, H.; Zheng, S.; Xiong, H.; Alwahibi, M.S.; Niu, X. Biosynthesis of copperoxide nanoparticles using Abies spectabilis plant extract and analyzing its antinociceptive and anti-inflammatory potency in various mice models. Arab. J. Chem. 2020, 13, 6995–7006. [Google Scholar] [CrossRef]
  73. Manasa, D.J.; Chandrashekar, K.R.; Kumar, D.M.; Niranjana, M.; Navada, K.M. Mussaenda frondosa L. mediated facile green synthesis of Copper oxide nanoparticles–Characterization, photocatalytic and their biological investigations. Arab. J. Chem. 2021, 14, 103184. [Google Scholar] [CrossRef]
  74. Yu, H.P.; Liu, F.C.; Lin, C.Y.; Umoro, A.; Trousil, J.; Hwang, T.L.; Fang, J.Y. Suppression of neutrophilic inflammation can be modulated by the droplet size of anti-inflammatory nanoemulsions. Nanomedicine 2020, 15, 773–791. [Google Scholar] [CrossRef] [PubMed]
  75. Rajeshkumar, S.; Menon, S.; Ponnanikajamideen, M.; Ali, D.; Arunachalam, K. Anti-inflammatory and antimicrobial potential of Cissus quadrangularis-assisted copper oxide nanoparticles. Jour. Nanomater. 2021, 2021, 1–11. [Google Scholar] [CrossRef]
  76. Madene, A.; Jacquot, M.; Scher, J.; Desobry, S. Flavour encapsulation and controlled release—A review. Int. J. Food Sci. Technol. 2006, 41, 1–21. [Google Scholar] [CrossRef]
  77. Kumar, M.; Bishnoi, R.S.; Shukla, A.K.; Jain, C.P. Techniques for Formulation of Nanoemulsion Drug Delivery System: A Review. Prev. Nutr. Food Sci. 2019, 24, 225–234. [Google Scholar] [CrossRef]
  78. Thakkar, H.P.; Khunt, A.; Dhande, R.D.; Patel, A.A. Formulation and evaluation of Itraconazole nanoemulsion for enhanced oral bioavailability. J. Microencapsul. 2015, 32, 559–569. [Google Scholar] [CrossRef]
  79. Mathiyalagan, R.; Wang, C.; Kim, Y.J.; Castro-Aceituno, V.; Ahn, S.; Subramaniyam, S.; Simu, S.Y.; Jiménez-Pérez, Z.E.; Yang, D.C.; Jung, S.-K. Preparation of polyethylene glycol-ginsenoside Rh1 and Rh2 conjugates and their efficacy against lung cancer and inflammation. Molecules 2019, 24, 4367. [Google Scholar] [CrossRef]
  80. Zhang, R.; Rupa, E.J.; Zheng, S.; Nahar, J.; Yang, D.C.; Kang, S.C.; Wang, Y. Panos-Fermented Extract-Mediated Nanoemulsion: Preparation, Characterization, and In Vitro Anti-Inflammatory Effects on RAW 264.7 Cells. Molecules 2022, 27, 218. [Google Scholar] [CrossRef]
  81. Wang, X.; Jiang, Y.; Wang, Y.W.; Huang, M.T.; Ho, C.T.; Huang, Q. Enhancing anti-inflammation activity of curcumin through O/W nanoemulsions. Food Chem. 2008, 108, 419–424. [Google Scholar] [CrossRef]
  82. Borges, R.S.; Keita, H.; Ortiz, B.L.S.; dos Santos Sampaio, T.I.; Ferreira, I.M.; Lima, E.S.; de Jesus Amazonas da Silva, M.; Fernandes, C.P.; de Faria Mota Oliveira, A.E.M.; da Conceição, E.C.; et al. Anti-inflammatory activity of nanoemulsions of essential oil from Rosmarinus officinalis L.: In vitro and in zebrafish studies. Inflammopharmacology 2018, 26, 1057–1080. [Google Scholar] [CrossRef]
  83. Tran, T.H.M.; Puja, A.M.; Kim, H.; Kim, Y.J. Nanoemulsions prepared from mountain ginseng-mediated gold nanoparticles and silydianin increase the anti-inflammatory effects by regulating NF-κB and MAPK signaling pathways. Biomater. Adv. 2022, 137, 212814. [Google Scholar] [CrossRef] [PubMed]
  84. Najda, A.; Bains, A.; Klepacka, J.; Chawla, P. Woodfordia fruticosa extract nanoemulsion: Influence of processing treatment on droplet size and its assessment for in vitro antimicrobial and anti-inflammatory activity. Front. Nutr. 2022, 9, 944856. [Google Scholar] [CrossRef]
  85. El-Naggar, M.E.; Hussein, J.; El-sayed, S.M.; Youssef, A.M.; El Bana, M.; Latif, Y.A.; Medhat, D. Protective effect of the functional yogurt based on Malva parviflora leaves extract nanoemulsion on acetic acid-induced ulcerative colitis in rats. J. Mater. Res. Technol. 2020, 9, 14500–14508. [Google Scholar] [CrossRef]
  86. Hoseny, S.S.; Soliman, A.M.; Fahmy, S.R.; Sadek, S.A. Development of a Novel Pomegranate Polysaccharide Nanoemulsion Formulation with Anti-Inflammatory, Antioxidant, and Antitumor Properties. Curr. Drug Deliv. 2022, 20, 575–586. [Google Scholar] [CrossRef]
  87. Rahimi Kalateh Shah Mohammad, G.; Homayouni Tabrizi, M.; 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, 30. [Google Scholar] [CrossRef]
  88. Kim, C.S.; Nguyen, H.D.; Ignacio, R.M.; Kim, J.H.; Cho, H.C.; Maeng, E.H.; Kim, Y.R.; Kim, M.K.; Park, B.K.; Kim, S.K. Immunotoxicity of zinc oxide nanoparticles with different size and electrostatic charge. Int. J. Nanomed. 2014, 9, 195–205. [Google Scholar] [CrossRef]
  89. Hooper, H.L.; Jurkschat, K.; Morgan, A.J.; Bailey, J.; Lawlor, A.J.; Spurgeon, D.J.; Svendsen, C. Comparative chronic toxicity of nanoparticulate and ionic zinc to the earthworm Eisenia veneta in a soil matrix. Environ. Int. 2011, 37, 1111–1117. [Google Scholar] [CrossRef] [PubMed]
  90. Jang, J.; Lim, D.H.; Choi, I.H. The impact of nanomaterials in immune system. Immune Netw. 2010, 10, 85–91. [Google Scholar] [CrossRef]
  91. Pasupuleti, S.; Alapati, S.; Ganapathy, S.; Anumolu, G.; Pully, N.R.; Prakhya, B.M. Toxicity of zinc oxide nanoparticles through oral route. Toxicol. Ind. Health 2012, 28, 675–686. [Google Scholar] [CrossRef] [PubMed]
  92. Naz, S.; Gul, A.; Zia, M. Toxicity of copper oxide nanoparticles: A review study. IET Nanobiotechnol. 2020, 14, 1–13. [Google Scholar] [CrossRef] [PubMed]
  93. McClements, D.J.; Rao, J. Food-grade nanoemulsions: Formulation, fabrication, properties, performance, biological fate, and potential toxicity. Crit. Rev. Food Sci. Nutr. 2011, 51, 285–330. [Google Scholar] [CrossRef] [PubMed]
  94. Henry, J.V.; Fryer, P.J.; Frith, W.J.; Norton, I.T. Emulsification mechanism and storage instabilities of hydrocarbon-in-water sub-micron emulsions stabilised with Tweens (20 and 80), Brij 96v and sucrose monoesters. J. Colloid Interface Sci. 2009, 338, 201–206. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bio formation and structure of phyto-metal nanoparticles and nanoemulsion.
Figure 1. Bio formation and structure of phyto-metal nanoparticles and nanoemulsion.
Immuno 03 00012 g001
Figure 2. Inflammatory mechanism of the metal nanoparticle.
Figure 2. Inflammatory mechanism of the metal nanoparticle.
Immuno 03 00012 g002
Figure 3. Inflammatory mechanism of medicinal plant-based nanoemulsion.
Figure 3. Inflammatory mechanism of medicinal plant-based nanoemulsion.
Immuno 03 00012 g003
Table 1. Anti-inflammatory effects of current gold and silver nanoparticles.
Table 1. Anti-inflammatory effects of current gold and silver nanoparticles.
NanoparticleMedicinal PlantsObservationCharacteristics
Size/Shape
Anti-Inflammatory ActionRef
GoldPanax gin-seng Meyer leafIn vitro10–20 nmReduction of the expression of the inflammatory mediators in the NF-κB signaling pathway[33]
GoldSuaeda japonica
Leaf
In vitro8.75 nm
Crystalline
Suppress the generation of
nitric oxide (NO) and repress the expression of the pro-inflammatory gene
[35]
GoldRosa ru-gosa(beach rosea)In vitro38.2 ± 3.7 nm
Polygonal
Treat skin inflammation by reducing oxidative stress via the MAPK and NF-κB sig-naling pathways[38]
GoldHypoxis hemerocallideaIn vitro26 ± 2 nm
Spherical
Reduce the amounts of pro-inflammatory cytokines in macrophage cells[41]
GoldHibiscus syriacus LIn vitro3–20 nm
Spherical
Suppress pro-inflammatory cytokines and decrease the expressions of PINK1 and Parkin in autophagy-dependent mechanism[46]
SilverAzadirachta indica kernelIn vitro19.27–22.15 nm
Spherical
Control protein degradation to fight inflammation[47]
SilverCotyledon orbiculataIn vitro20 to 40 nmSuppress the secretion of the pro-inflammatory marker in LPS-treated macrophage[48]
SilverCin-namomum zeylanicum barkIn vitro60–80 nm
Spherical
Decrease TNF-α, IL-6, and IL-18 inflammatory markers of PCOS[49]
SilverSyzygium cumini fruitIn vitro~47 nm
Spherical
Protein denaturation in higher concentration[50]
SilverSelaginella myosurusIn vitro33.7, 44.2Inhibition of thermally induced denaturation of albumin[51]
Table 2. Anti-inflammatory effects of current copper and zinc nanoparticles.
Table 2. Anti-inflammatory effects of current copper and zinc nanoparticles.
NanoparticleMedicinal PlantsObservationCharacteristics
Size/Shape
Anti-Inflammatory ActionRef
ZnTerminalia ferdinandiana (Kakadu plum)In vitro21.89 nm
Crystalline
Inhibition of pro-inflammatory nitric oxide production[63]
ZnZingiber officinaleIn vitro30 nm
Spherical
Inhibition of COX1 and COX2[66]
ZnPolygala tenuifolia root)In vitro9.22 nm
Spherical
Downregulation of both mRNA and protein ex-pressions of inflammatory mediators[68]
ZnKalanchoe pinnataIn vitro24 nmSuppress pro-inflammatory mediators such as interleukin 6 (IL-6), interleukin 1 (IL-1), tumor necrosis factor (TNF-α), and cyclooxy-genase-2 (COX-2)[69]
CopperMyrtus Communis leavesIn vitro53.55 nm
Crystalline
Inhibition of protein oxidation[70]
CopperMucuna pruriens seedIn vitroNASuppress the inflammatory mediators[71]
CopperAbies spectabilisIn vitroNASuppress the inflammatory cytokines IL-1β, IL-6, and TNF-α[72]
Table 3. Medicinal plant-based nanoemulsions (O/W) and their anti-inflammatory activities.
Table 3. Medicinal plant-based nanoemulsions (O/W) and their anti-inflammatory activities.
Medicinal PlantsPhasesTypesObservationMechanism/Pathway/ActionRef
Panax ginseng leaf extractLeaf extract + Water + Sea buckthorn oilO/WIn vitroSuppression of pro-inflammatory mediators for (Cox 2, IL-6, IL-1β, and TNF-α, NF-κB, Ikkα, and iNOS) gene expression[80]
CurcuminCurcumin + Water + MCTO/WIn vitroInhibition of TPA-induced edema of mouse ear[81]
Rosmarinus officinalis LLeaf extract + Essential oilO/WIn vitroInhibiting the production of the pro-inflammatory mediator nitric oxide[82]
Mountain gin-sengGinseng ex-tract + Gin-seng seed oilO/WIn vitroInhibition of pro-inflammatory genes and proteins, including IL-1β, IL-6, and TNF-α via
NF-κB and MAPK signaling pathways
[83]
Woodfordia fruticosa flower extractFlower extract + Sunflower seed oilO/WBacterial cell membraneInhibit the release of inflammatory mediators and stabilize cell membrane[84]
Malva parviflora leaf extractLeaf extract + Yoghurt bO/WIn vitroDiminish the production of superoxide by inhibiting NADPH oxidase[85]
Punica granatum peelFruit Peel extract + Cremophor RH40O/WIn vitroStabilize the lysosomal membrane as anti-inflammatory efficacy.[86]
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

Puja, A.M.; Rupa, E.J.; Kim, Y.J.; Yang, D.-C. Medicinal Plant Enriched Metal Nanoparticles and Nanoemulsion for Inflammation Treatment: A Narrative Review on Current Status and Future Perspective. Immuno 2023, 3, 182-194. https://doi.org/10.3390/immuno3020012

AMA Style

Puja AM, Rupa EJ, Kim YJ, Yang D-C. Medicinal Plant Enriched Metal Nanoparticles and Nanoemulsion for Inflammation Treatment: A Narrative Review on Current Status and Future Perspective. Immuno. 2023; 3(2):182-194. https://doi.org/10.3390/immuno3020012

Chicago/Turabian Style

Puja, Aditi Mitra, Eshrat Jahan Rupa, Yeon Ju Kim, and Deok-Chun Yang. 2023. "Medicinal Plant Enriched Metal Nanoparticles and Nanoemulsion for Inflammation Treatment: A Narrative Review on Current Status and Future Perspective" Immuno 3, no. 2: 182-194. https://doi.org/10.3390/immuno3020012

Article Metrics

Back to TopTop