Recent progress in nano-TCM active ingredient co-delivery systems for

Introduction

Inflammation is an essential protective response of the immune system to harmful stimuli, such as allergens or tissue injury. However, the uncontrolled activation of inflammation, driven by an overproduction of pro-inflammatory mediators and oxidative stress, is a major factor contributing to tissue damage and the development of various chronic diseases.1 Inflammatory cells release reactive oxygen species (ROS), pro-inflammatory cytokines, transcription factors, and chemokines, which further amplify oxidative stress and disrupt the extracellular matrix, ultimately exacerbating the inflammatory response.2,3 A growing body of evidence supports the therapeutic potential of targeting inflammation to alleviate the progression of a wide range of diseases.4

Although synthetic anti-inflammatory drugs are commonly prescribed, their use is often accompanied by significant adverse effects, raising concerns about their long-term safety. Nonsteroidal anti-inflammatory drugs (NSAIDs), the most widely used class of anti-inflammatory medications, are associated with serious gastrointestinal complications, including erosions, ulcers, bleeding, and perforations, affecting 1–2% of patients after three months of therapy.5 Furthermore, NSAIDs have been linked to the development of small intestinal strictures, and aspirin, in particular, may induce platelet apoptosis, leading to thrombocytopenia. These issues highlight the limitations of conventional anti-inflammatory therapies and underscore the need for safer alternatives.6 In contrast, TCM, with its rich array of bioactive compounds derived from natural sources, offers a promising alternative for the treatment of inflammatory diseases. TCM has garnered increasing attention in recent years, owing to its diverse pharmacological activities, including anti-inflammatory, anti-tumor, and immune-regulatory effects.7 Many active ingredients from TCM can simultaneously target multiple molecular pathways, thereby enhancing therapeutic efficacy and modulating immune responses in a more comprehensive manner.8–10 Despite its potential, the clinical application of TCM is limited by challenges such as poor solubility, unstable pharmacokinetics, non-specific side effects, and limited biofilm penetration of the active compounds. These obstacles have hindered the full therapeutic potential of TCM, highlighting the need for innovative approaches to improve the delivery and efficacy of TCM-based therapies.11,12

To improve the utilization of TCM, researchers have focused on the medical applications of nanocarriers. TCM has long been delivered through decoctions, pills, powders, pastes, and tinctures. However, these delivery methods often result in low absorption efficiency (eg, for polysaccharides and proteins), poor stability (prone to degradation by light and heat), and weak targeting (resulting in systemic distribution that may cause side effects in non-target organs). Additionally, the complex composition of TCM formulas delivered by traditional methods poses challenges in quality control.13,14 In comparison to conventional drug delivery systems, nano-based TCM co-delivery systems offer superior safety and efficacy. These systems benefit from the inherent anti-inflammatory and antioxidant properties of the nanocarriers, as well as their specific physical/chemical targeting, and enhanced biocompatibility.15,16 Lipid-based nanocarriers have gained attention due to their amphiphilicity, high stability, and excellent biocompatibility.17 Polymeric nanoparticles have been widely studied for their high encapsulation efficiency.18 Biomimetic nanocarriers, such as cell-based nanocarriers and exosomes, are recognized for their low immunogenicity, extended in vivo circulation time, and strong targeting ability.19 Our review analyzes the anti-inflammatory mechanisms of various active components in TCM, introduces the classifications and properties of nanocarriers, and highlights the latest advancements in TCM delivery systems for treating inflammation-mediated diseases. It also discusses the advantages and unresolved issues of nanocarrier-based delivery of TCM, offering new perspectives for the development of this field.

Advantages and Limitations of TCM Active Ingredients in the Treatment of Inflammation-Mediated Diseases

Resveratrol

Resveratrol, a natural polyphenolic compound, demonstrates a wide range of biological activities, including free radical scavenging, regulation of antioxidant enzyme expression and activity, anti-inflammatory, anti-aging, anti-glycation, anti-cancer, neuroprotective, and cardioprotective effects.20,21 In high-fat diet-induced obesity (DIO) mice, resveratrol exhibits potent anti-inflammatory and antioxidant properties,22 alleviating allergic asthma by inhibiting JNK and NF-κB signaling pathways.23 Furthermore, resveratrol effectively reduces insulin resistance and macrophage infiltration in adipose tissue, improving inflammation in both the peripheral and central nervous systems of DIO mice.24 Recent studies have shown that the anti-inflammatory effects of resveratrol are linked to SIRT1 activation. For instance, resveratrol modulates the SIRT1/NF-κB signaling pathway to suppress the expression of pro-inflammatory factors such as COX-2, IL-1, and IL-6, thereby alleviating colitis.25 Additionally, resveratrol has demonstrated therapeutic effects on osteoarthritis (OA) and vasculitis.26,27 Despite its high absorption rate, the low solubility, poor stability, short half-life, and rapid metabolism of resveratrol contribute to its low bioavailability, which limits its clinical application.28

Quercetin

Quercetin, a polyphenolic flavonoid predominantly found in fruits and vegetables, exhibits anti-inflammatory, antioxidant, and autophagy-inducing properties. Consequently, it has been investigated for the prevention and treatment of cancer, cardiovascular diseases, chronic inflammation, oxidative stress, and neurodegenerative disorders.29 In vitro studies demonstrate that quercetin inhibits TNF-α-induced inflammation in macrophages and adipocytes by modulating MAPK (JNK and ERK) and nuclear factor-kappa B(NF-κB) pathways.30 In vivo, it alleviates experimentally induced hepatitis and interstitial nephritis by suppressing macrophage M1 polarization.31,32 In OA animal models, quercetin acting as a SIRT1 agonist activates the AMPK signaling pathway to suppress inflammation and apoptosis in chondrocytes,33 while promoting cartilage repair through M2 polarization of synovial macrophages.34 Conversely, quercetin has also been shown to reduce excessive extracellular matrix accumulation in renal interstitial cells by inhibiting M2 polarization via antagonism of the TGF-β1/Smad2/3 signaling pathway.31 Given these complex effects on macrophage polarization, further studies are required to elucidate its precise mechanisms of action. Additionally, quercetin significantly reduces plasma histamine levels and serum IgE concentrations, thereby alleviating peanut-induced allergic responses in rats.35 However, its high first-pass elimination rate—resulting in rapid excretion within 24 hours—and inherent hydrophobicity limit its bioavailability and therapeutic potential.36,37

Curcumin

Curcumin, a polyphenolic compound extracted from the rhizome of Curcuma longa (turmeric), exhibits regulatory effects on inflammation, oxidative stress, and various cellular processes including proliferation, differentiation, and survival.38 Recent studies have demonstrated that the anti-inflammatory properties of curcumin are primarily mediated through the modulation of several key signaling pathways, such as NF-κB, peroxisome proliferator-activated receptor gamma (PPAR-γ), and Toll-like receptor (TLR)/myeloid differentiation protein 2 (MD2) pathways. For example, curcumin mitigates colitis and rheumatoid arthritis by inhibiting IκB kinase (IKK) activity and IκB-α phosphorylation, thereby blocking NF-κB pathway activation,39,40 In addition, upregulation of PPAR-γ expression has been shown to attenuate inflammation and reduce ROS production.41 Acting as a PPAR-γ agonist, curcumin suppresses angiotensin II–induced inflammation in vascular smooth muscle cells.42 Moreover, aberrant activation of TLR signaling complexes serves as an upstream trigger of inflammatory responses. Curcumin and its analogs competitively bind to MD2, thereby inhibiting the TLR4-MD2 complex and reducing the secretion of pro-inflammatory cytokines. This mechanism has been implicated in the amelioration of acute lung injury (ALI) and sepsis.43 Furthermore, curcumin modulates macrophage polarization by inhibiting M1 phenotypes while promoting M2 polarization, contributing to its immunomodulatory effects.44,45 Despite its broad therapeutic potential, the clinical application of curcumin remains limited due to its low water solubility, poor bioavailability, rapid metabolic degradation, and fast systemic elimination.46

Honokiol

Honokiol is a natural polyphenolic compound extracted from the bark and leaves of Magnolia species belonging to the Magnoliaceae family. It exhibits low toxicity and a broad spectrum of biological activities, including anti-inflammatory, antioxidant, anti-tumor, anti-obesity, and neuroprotective effects.47 The anti-inflammatory effects of honokiol are closely associated with the activation of SIRT3, a mitochondrial deacetylase that plays a crucial role in maintaining mitochondrial function and redox homeostasis.48,49 For instance, honokiol alleviates osteoarthritis by targeting the SIRT3–COX4I2 axis, thereby reprogramming mitochondrial respiratory chain complexes.48 Additionally, honokiol regulates the differentiation of T helper 17 (Th17) cells by activating SIRT3, which in turn inhibits the STAT3/RORγt signaling pathway, leading to decreased expression of IL-17 and interleukin-21 (IL-21), and ultimately reducing intestinal inflammation in colitis models.50 Furthermore, honokiol exerts anti-colitic effects through modulation of the PPARγ/NF-κB, AMP-activated protein kinase (AMPK), and nuclear factor erythroid 2–related factor 2/heme oxygenase-1 (NRF2/HO-1) signaling pathways.51 Recent findings also suggest that honokiol can activate SIRT1, expanding its regulatory effects on cellular stress responses.52 Moreover, honokiol has been shown to attenuate lung inflammation by regulating AMPK and superoxide dismutase 2 (SOD2) signaling pathways.53,54 Taken together, these findings highlight honokiol as a promising therapeutic candidate for inflammation-related diseases. However, its clinical application is substantially hindered by poor physicochemical properties, including low aqueous solubility, instability, and rapid metabolic degradation. These factors result in an oral bioavailability of approximately 5%, significantly limiting its therapeutic potential.55,56

Bergenin

Bergenin, an isocoumarin compound derived from Bergenia species, exhibits diverse biological activities, including anti-inflammatory, antioxidant, antiviral, antifungal, and organ-protective effects.57 As a SIRT1 agonist, bergenin alleviates asthma by modulating the NF-κB pathway in macrophages, thereby suppressing the expression of IL-1β, IL-5, IL-6, and MMP-9.58 Additionally, it mitigates OA by regulating macrophage M1/M2 polarization.59 PPAR-γ, an upstream regulator of SIRT1, can be activated by bergenin, leading to upregulated SIRT1 expression. This mechanism reduces oxidative stress and inflammation, offering therapeutic benefits for colitis and Alzheimer’s disease-related dementia.60,61 Furthermore, bergenin’s anti-inflammatory effects are associated with the PI3K/AKT signaling pathway.62 However, its clinical application is hindered by low water solubility, poor permeability, and limited biofilm penetration,63 as well as instability and susceptibility to degradation in neutral and alkaline environments.64 These limitations contribute to its low bioavailability, necessitating higher doses to achieve effective therapeutic blood concentrations.65

Ginsenosides

Ginsenosides (G), the primary bioactive constituents of ginseng, regulate diverse cellular processes by interacting with cell membranes, kinases, and transcription factors.66 Specifically, G-Rb1, G-Rb2, G-Rd, G-Re, G-Rg1, G-Rg3, Rh1, and compound K (CK) exhibit potent anti-inflammatory effects in vivo and in vitro by suppressing the activity of IL-1 receptor-associated kinase (IRAK-1), NF-κB, and MAPK signaling pathways.67–73 Notably, G-Rb2,74 G-Rc,75 G-Rg1,76 G-Rg3 exert anti-inflammatory actions via SIRT1 activation.77 For example, G-Rg3 mitigates microglial inflammation by modulating the SIRT1/NRF2/NF-κB and AMPK/PI3K/AKT pathways.78 Given that mitochondrial dysfunction is a key driver of inflammation,79 G-Rg3 further ameliorates inflammatory responses by enhancing mitochondrial biogenesis through AMPK-mediated mitophagy and upregulation of PGC-1α and related genes.80,81 Additionally, G-Rg6 suppresses TLR4-mediated systemic inflammation,82 while G-Rh1 modulates the ERK and STAT (STAT1/STAT3) pathways to attenuate inflammation.83 However, the therapeutic potential of ginsenosides is limited by their susceptibility to degradation or transformation in gastric acid and intestinal bacteria, with metabolites potentially inhibiting the absorption of parent compounds. Although their lipophilicity aids biofilm penetration, poor oral absorption and low bioavailability remain major challenges (Figure 1).66,84

Figure 1 The anti-inflammatory molecular mechanisms of TCM active ingredients. Resveratrol, quercetin, curcumin, bergenin, and ginsenosides exert anti-inflammatory effects by targeting membrane receptors (TLR4, MD2), nuclear regulators (PPARγ, SIRT1), and AMPK, thereby suppressing pro-inflammatory NF-κB, MAPK, and PI3K/AKT signaling pathways while mitigating oxidative stress and promoting mitochondrial biogenesis. Additionally, honokiol, as a SIRT3 agonist, modulates immune responses by inhibiting Th17 cell differentiation through suppression of the STAT3/RORγt pathway.

Berberine

Berberine (BBR), an isoquinoline alkaloid derived from various medicinal plants, exhibits potent anti-inflammatory properties. BBR and its derivatives demonstrate therapeutic efficacy in inflammatory diseases affecting the gut, lungs, skin, and bone.85,86 Mechanistically, BBR suppresses the expression of pro-inflammatory genes (eg, IL-1β, IL-6, and iNOS) by activating the AMPK signaling pathway and inhibiting MAPKs phosphorylation in macrophages.87 Its anti-inflammatory effects further involve modulation of the Nrf2 and NF-κB pathways.88 Notably, BBR significantly inhibits IgE production, highlighting its potential in managing food allergies. This immunomodulatory effect is linked to BBR’s ability to suppress IκB phosphorylation and regulate gut microbiota composition.89,90 Despite demonstrating multiple beneficial pharmacological activities against various diseases, the therapeutic application of berberine is substantially limited by several pharmacokinetic challenges, including poor oral bioavailability, low gastrointestinal absorption, and extensive first-pass elimination.91,92

Advantages of Nano-TCM Active Ingredient Co-Delivery Systems

Improve the Bioavailability of TCM Active Ingredients

Most TCM preparations suffer from poor water solubility, chemical instability, rapid first-pass metabolism, and limited biofilm penetration, all of which significantly compromise their oral bioavailability.17 These pharmacokinetic limitations present major challenges for clinical translation of TCM therapies.

Recent advances in nanomedicine have demonstrated that nanocarriers can effectively overcome these delivery challenges. Engineered nanocarriers enhance the solubility, stability, and absorption efficiency of TCM active ingredients while simultaneously reducing their potential toxicity. Compared to free TCM compounds, nano-TCM co-delivery systems show significantly improved bioavailability and enhanced therapeutic effects, including superior anti-inflammatory and antioxidant activities.93–96 Notably, certain nanocarriers such as nanoemulsions and dendrimers exhibit exceptional biofilm penetration capabilities through fusion or permeation mechanisms.97,98 Biomimetic nanocarriers, characterized by their low immunogenicity and high biocompatibility, show particular promise for overcoming biological barriers and avoiding immune system clearance.99 Furthermore, these nano-TCM delivery platforms offer flexible administration routes, including intravenous, oral, transdermal, and various mucosal (ocular, nasal) delivery options.100,101

Enhance Drug Delivery Targeting Inflammation

Extensive research has confirmed that nanocarriers can significantly alter drug biodistribution patterns.102,103 A key advantage of nanocarriers is their preferential accumulation in inflamed tissues. For example, cationic liposomes demonstrate significantly higher accumulation in lung tissues of inflamed animal models compared to healthy controls.104 This enhanced permeability and retention (EPR) effect, mediated by pathophysiological vascular leakage, represents the passive targeting mechanism of nanocarriers.105 Beyond passive targeting, nanocarriers can be engineered for active targeting of specific molecules or cellular phenotypes within inflammatory microenvironments. Liposomes, for instance, can be designed to target specific macrophage phenotypes, thereby modulating phagocytic activity and cytokine secretion profiles.106 Macrophage-derived extracellular vesicles show particular promise for neuroinflammation therapy due to their enhanced blood-brain barrier (BBB) penetration capabilities.107 Surface modification strategies further expand nanocarrier targeting potential. Functionalization with surfactants, antibodies, polymers (synthetic or natural), silicon dioxide, metals, or other materials can precisely enhance targeting specificity.108 Galactose and folic acid-modified resveratrol, which shows improved intestinal absorption and transcellular transport, enhancing its anti-inflammatory efficacy both in vitro and in vivo.109,110 Lactoferrin-conjugated resveratrol, demonstrating enhanced BBB penetration and neuroprotective effects.111 Mannose-modified albumin, which effectively targets mannose receptors overexpressed on inflammatory cells.112

Control Drug Release

Many TCM active ingredients suffer from poor water solubility, short half-lives, and rapid first-pass elimination. Patients frequently require high-dose regimens to attain therapeutic efficacy, decreasing medication adherence while increasing adverse event risks.17,113,114 Nanocarrier-based delivery systems have emerged as a promising solution to overcome these limitations. Recent studies demonstrate that engineered nanocarriers can significantly prolong drug release profiles. For example, Silica nanoparticles enable sustained release of silymarin for up to 72 hours.115 Liposome-encapsulated cryptotanidone administered every 48 hours shows comparable efficacy to daily pirfenidone dosing.116 Advanced nanocarrier systems can be designed with stimulus-responsive properties for site-specific drug release. Conventional liposomes maintain stability under physiological conditions but release payloads upon encountering specific internal/external triggers. Functionalized liposomes incorporating thermosensitive or pH-sensitive polymers enable precise control of drug release in response to local microenvironment changes.117 Magnetic nanoparticles have also been investigated for controlled drug release under external magnetic field influence.108 These controlled-release strategies provide multiple therapeutic advantages, including minimization of direct drug-mucosa contact, prevention of mucosal irritation caused by local drug accumulation, maintenance of optimal therapeutic drug concentrations, reduction in dosing frequency.17,118

As Carriers to Co-Deliver Active Ingredients or Therapeutic Agents to Enhance Efficacy

Nanocarrier-mediated co-delivery systems leverage the principle of pharmacological synergy to simultaneously transport multiple TCM active ingredients or therapeutic agents. This strategy enables concurrent modulation of diverse molecular pathways while minimizing toxicity and adverse effects through reduced dosage requirements. Curcumin serves as an exemplary combination partner due to its dual capacity to inhibit drug resistance-associated transcription factor activation and downregulate drug transporter activity.119 For instance, Curcumin-quercetin nanoemulsions showing superior antiviral efficacy with reduced doses and improved targeting specificity compared to monotherapies.120 Macrophage membrane-coated curcumin-platycodin systems achieving significant anti-inflammatory effects in ALI murine models (Figure 2).121

Figure 2 Nanocarrier systems offer significant advantages for delivering TCM active ingredients. The clinical translation of TCM active ingredients is often hindered by inherent limitations, including poor solubility, low stability, rapid clearance, unfavorable pharmacokinetics, and nonspecific side effects—all contributing to low bioavailability. Functional benefits of nanocarriers, such as intrinsic anti-inflammatory/antioxidant properties, enhanced solubility, controlled release, targeted delivery, and co-delivery of therapeutic agents, position them as powerful platforms for TCM delivery.

Classification of Nanocarriers

Exogenous Nanocarriers

Lipid Nanocarriers

Liposomes

Liposomes are stable spherical vesicles with a bilayer structure formed by phospholipids and cholesterol. Their unique amphiphilic nature, combined with high biocompatibility, structural stability, and ease of surface modification, makes them particularly valuable for overcoming the delivery challenges of TCM compounds.17 Numerous studies have demonstrated the effectiveness of liposomes for encapsulating and delivering TCM bioactive components including quercetin, resveratrol, and curcumin.122–124

Solid Lipid Nanoparticles (SLNs)

SLNs represent another important class of nanocarriers, featuring a solid lipid core that maintains crystalline structure at room temperature. These nanoparticles offer several advantages over liposomes, including greater physical stability and lower production costs.125 A distinctive characteristic of SLNs is their tendency to adsorb drugs primarily on the particle surface rather than encapsulating them within the core.126 This property, along with their ability to load both hydrophilic and lipophilic drugs, provide targeted delivery, protect payloads from degradation, and enable controlled release, makes SLNs attractive for TCM applications.17 However, a significant limitation arises during preparation – the formation of highly crystalline structures in the lipid matrix can lead to drug expulsion and consequently reduced drug loading capacity.127

Nanostructured Lipid Carriers (NLCs)

To address these limitations of SLNs, researchers developed NLCs by incorporating liquid lipids (oils) into the solid matrix. This modification disrupts the perfect crystalline structure of SLNs, resulting in improved nanoparticle stability and minimized drug leakage.125 NLCs have shown particular promise for delivering various TCM compounds such as bergenin,128 quercetin,129 and ginsenosides.130

Nanoemulsions

Nanoemulsions, consisting of two immiscible liquid phases stabilized by surfactants and co-surfactants, offer another promising delivery platform. These systems are thermodynamically stable and can maintain their stability for extended periods (up to 3 months) at room temperature.105 The large interfacial surface area of nanoemulsions facilitates efficient drug release and absorption. Furthermore, their versatility allows administration through multiple routes including oral, topical, and parenteral delivery.100

Polymeric Nanocarriers

Polymeric Micelles

Polymeric micelles represent an important class of nanocarriers characterized by their core-shell architecture, formed through the self-assembly of amphiphilic block copolymers.131 The hydrophobic core serves as an effective compartment for encapsulating hydrophobic drugs through either covalent conjugation or physical entrapment, providing protection against degradation while significantly improving drug solubility. Meanwhile, the hydrophilic shell contributes several advantageous features, including excellent biocompatibility, prolonged blood circulation time,18 enhanced permeability, active targeting capability, and stimuli-responsive properties to temperature and pH changes.17,28 With their relatively small size range (10–200 nm), superior solubilization capacity, and straightforward preparation and sterilization processes, polymeric micelles offer distinct advantages over other nanocarrier systems. However, their clinical translation faces challenges due to stability limitations that require further optimization.131

Polymeric Nanoparticles

Polymeric nanoparticles are submicron-sized colloidal particles fabricated from either natural or synthetic polymers such as Poly(lactic-co-glycolic acid) (PLGA), Polylactide (PLA), silk fibroin (SF), chitosan, inulin, and gelatin. These nanoparticles share many beneficial properties with polymeric micelles, including both active and passive targeting mechanisms, as well as temperature- and pH-responsive behaviors.132 Among these materials, PLGA has emerged as one of the most versatile and widely investigated polymers in nanomedicine applications.

Dendrimers

Dendrimers constitute a unique class of highly branched, monodisperse macromolecules with well-defined core-shell nanostructures. Their distinctive architecture features discrete dendritic branches that radiate outward in concentric layers from a central core, creating abundant surface functional groups for drug conjugation or chemical modification, along with internal cavities for drug encapsulation.133 The precise control over dendrimer synthesis allows for tailored adjustment of molecular weight, surface functionality, and hydrophilicity, enabling optimization of drug-loading capacity, biocompatibility, and pharmacokinetic profiles.134 Furthermore, dendrimers offer administration route flexibility,101 making them particularly attractive for delivering both small molecule drugs and larger biomolecules. Nevertheless, potential cytotoxicity associated with cationic surface charges remains an important consideration that requires careful evaluation during dendrimer design.133

Inorganic Nanoparticles

Inorganic nanoparticles are nanoparticles prepared from inorganic materials such as metals, oxides, semiconductors and carbon-based structures. Owing to their excellent physical stability, large surface area, and sensitivity to light, magnetic fields, and ultrasonic signals, inorganic nanoparticles facilitate drug delivery through light/heat and magnetic field therapy.135 At present, widely used inorganic nanoparticles include gold nanoparticles, silica nanoparticles, magnetic nanoparticles, and nanotubes.17,136

Nanocrystals

Drug nanocrystals represent a unique class of nanocarriers consisting of pure drug particles with diameters below 1 μm, typically stabilized in nanosuspension formulations. These nanocrystals can be prepared through two principal methodologies: top-down approaches involving mechanical size reduction techniques such as high-pressure homogenization or milling, and bottom-up approaches utilizing antisolvent crystallization processes. In the latter method, carefully selected antisolvents (including various surfactants and polymers) are employed to induce controlled solute supersaturation and subsequent crystallization. The exceptionally high surface area-to-volume ratio of nanocrystals constitutes their most distinctive physicochemical characteristic, which directly contributes to enhanced dissolution rates and improved bioavailability. However, due to their prolonged persistence in biological environments, factors such as nanotoxicity and dose must be considered during their interaction with biological tissues.137

Heparin Based Nanocarriers

Heparin, a naturally occurring glycosaminoglycan, exhibits remarkable biocompatibility and contains abundant chemically modifiable functional groups along its backbone.138 The low-molecular-weight derivatives of heparin (LMWHs) demonstrate enhanced multifunctionality, including anticoagulant activity, anti-inflammatory effects, angiogenesis inhibition, and antitumor properties.139 These unique characteristics have positioned heparin-based nanomaterials as promising drug delivery platforms. For instance, LMWH-conjugated polymeric micelles have shown significant potential in reducing atherogenesis in murine models by simultaneously addressing vascular inflammation and dyslipidemia.140 Additionally, heparin’s specific binding affinity for heparanase enables effective regulation of the PI3K/AKT/mTOR signaling pathway,141 a critical cascade involved in mediating inflammatory processes.142

Bionic Nanocarriers

Protein-Based Nanoparticles

Serum proteins represent a class of naturally occurring biomacromolecules that possess several advantageous characteristics for drug delivery applications, including exceptional biocompatibility, inherent biodegradability, intrinsic targeting capabilities, minimal toxicity, and low immunogenicity. These proteins contain abundant surface-exposed functional residues such as amino and carboxyl groups, which facilitate chemical conjugation with therapeutic agents and imaging probes. Among the most extensively studied serum protein-based nanocarriers are albumin, ferritin/apoferritin, transferrin, low-density lipoprotein, high-density lipoprotein, and hemoglobin, each demonstrating unique advantages for targeted drug delivery.143,144

Red Blood Cell (RBC)-Based Nanocarriers

As the most abundant circulatory cells, RBCs are easily accessible and exhibit several advantageous biological characteristics, including prolonged circulation time, low immunogenicity, and high deformability.145 Their natural ability to freely traverse vascularized tissues makes them ideal candidates for drug delivery applications. Recent studies have shown that functional modification of RBC membranes can significantly enhance their targeting specificity and controlled drug release capabilities.146 Currently, three main approaches are employed for drug loading onto RBCs: drug internalization through hypotonic preswelling techniques;147 drug anchoring on RBC membrane surfaces via covalent or non-covalent coupling methods;145 and drug encapsulation within membrane-derived vesicles.148 Notably, research has demonstrated that selective placement of intravascular catheters combined with intravenous injection of RBC-drug co-delivery systems can significantly enhance drug uptake in downstream target organs.149 However, this delivery system has limitations – RBCs cannot actively deliver drugs to extravascular targets.150 And the loaded drugs may disrupt normal RBC function, potentially leading to hemolysis and cellular aggregation.151,152

Macrophage-Based Nanocarriers

Macrophages, as natural immune cells, possess inherent anti-inflammatory properties through their phagocytic activity and innate inflammatory homing capabilities, making them promising cellular carriers for anti-inflammatory therapy. Current research has identified two primary approaches for drug loading into macrophages. The first approach relies on macrophage-mediated phagocytosis to internalize therapeutic agents.153 However, this method faces challenges due to potential drug cytotoxicity to macrophages and drug degradation by lysosomal enzymes. To overcome these limitations, researchers have developed strategies where drugs are first encapsulated within protective nanocarriers such as liposomes or polymeric nanoparticles before being loaded into macrophages.154,155 The alternative approach involves surface conjugation of drugs or drug-loaded nanoparticles to macrophages through covalent or non-covalent interactions.153 While this method avoids lysosomal degradation, it may interfere with critical cellular functions including signaling pathways and adhesion mechanisms.156

Exosomes

Exosomes are nanoscale extracellular vesicles (40–160 nm in diameter) secreted by nearly all cell types. These natural vesicles contain a diverse cargo of biologically active components, including proteins, nucleic acids, lipids, cytokines, enzymes, signal transduction proteins, and adhesion molecules.157 Their endogenous composition confers several advantageous properties for drug delivery, such as low toxicity, high biocompatibility, intrinsic targeting ability, and the capacity to cross biological barriers while avoiding immune clearance. However, significant challenges remain in the clinical translation of exosome-based therapies, primarily due to the inefficient isolation from biological fluids and cell culture media, as well as their inherent heterogeneity and complex composition.99 Interestingly, recent studies have identified milk as a promising alternative source of exosomes, as these vesicles demonstrate cross-species tolerance and minimal immunogenicity or inflammatory responses (Figure 3).158

Figure 3 Classification of nanocarriers and possible drug loading methods. This figure summarizes two major categories of nanocarriers (exogenous and bionic systems) for delivering TCM active ingredients, along with surface modification strategies (antibodies, peptides, monosaccharides, polysaccharides, folic acid, and ligands) to enhance targeting capability in co-delivery systems.

Nano-TCM Active Ingredient Co-Delivery Systems in Inflammation-Mediated Diseases

Neuroinflammatory Diseases

The BBB presents a significant challenge for drug delivery to the central nervous system, particularly for TCM active ingredients.159 To overcome this limitation, various nano-TCM delivery systems have been developed to enhance neuroprotective efficacy.

Research demonstrates that resveratrol-loaded PLGA and Polyethylene glycol (PEG) nanoparticles effectively cross the BBB, attenuating neuroinflammation and oxidative stress while providing neuroprotection.111,160 Similarly, exosome-encapsulated resveratrol exhibits potent anti-inflammatory effects in both central and peripheral nervous systems.107 These therapeutic benefits appear mediated through modulation of critical signaling pathways including STAT3, NF-κB, MAPK, and AKT.161,162

In Alzheimer’s disease (AD) pathology, characterized by extracellular amyloid-β (Aβ) aggregation.163 Nanocarrier systems have shown particular promise. Polymeric and silica nanoparticles significantly improve quercetin bioavailability, reducing neuronal apoptosis and Aβ plaque formation in AD models.164,165 Plasma exosome-delivered quercetin similarly ameliorates cognitive deficits in AD mice.166

Curcumin nanoformulations demonstrate multimodal neuroprotection by inhibiting M1 microglial activation, suppressing pro-inflammatory cytokines (TNF-α, IL-1β), and preserving BBB integrity during cerebral ischemia-reperfusion injury.167

Honokiol delivery systems exhibit excellent oral bioavailability and potent anti-AD effects, including reduced neuroinflammation (TNF-α, IL-6, IL-1β), suppressed glial activation, and decreased Aβ deposition.168 Intravenous honokiol nanocapsules additionally show efficacy in autoimmune encephalomyelitis models.169

While ginsenosides possess inherent neuroprotective properties, their poor BBB penetration limits clinical application.170 Innovative solutions include transferrin receptor-targeted nanoparticles for sustained Rg1 release in diabetic cerebral infarction,171 and macrophage membrane-coated G-Rg3 delivery systems that enhance brain targeting in ischemic injury (Table 1).172,173

Table 1 Nano-TCM Active Ingredient Co-Delivery Systems for Neuroinflammatory Diseases

Inflammatory Lung Disease

The lipid core nanocapsule-resveratrol co-delivery system demonstrates remarkable stability, enhanced oral bioavailability, and preferential lung accumulation. This formulation effectively mitigates ALI in murine models by suppressing ERK and PI3K/AKT pathway activation, thereby reducing both inflammatory responses and oxidative stress.174 Further enhancing targeting precision, folate-modified exosomes have been engineered to specifically deliver resveratrol and celastrol to M1 macrophages. This approach significantly inhibits M1 polarization in pulmonary macrophages, attenuates cytokine storm severity, and improves survival rates in septic mice.175

Macrophages’ innate capacity for inflammation-directed phagocytosis enables their effective accumulation in inflamed tissues. When employed as quercetin carriers, macrophages demonstrate significant efficacy in alleviating acute pneumonia in murine models.176 An alternative strategy utilizing nanoemulsions co-encapsulating quercetin and curcumin achieves a 99% viral inhibition rate following intranasal administration.120

Advanced targeting has been achieved through receptor-binding peptide (RBP)-modified exosomes, which selectively deliver curcumin to inflamed lung tissues.177 In ovalbumin (OVA)-induced asthma models, curcumin-loaded nanomicelles exhibit 9.24-fold greater oral bioavailability than free curcumin, accompanied by significantly enhanced anti-inflammatory effects.178

The bovine serum albumin-bergenin co-delivery system displays exceptional stability while effectively ameliorating ALI through multiple mechanisms: reducing pulmonary inflammatory cell infiltration, suppressing TNF-α production, and enhancing myeloperoxidase activity.179

For COVID-19-related pathologies, the PEG-albumin-ginsenoside system improves survival in animal models by concurrently inhibiting thrombosis, vasculitis, and cytokine storms via modulation of NF-κB and SREBP2 signaling pathways.180

Innovative cell membrane-based approaches have utilized MLE-12 pulmonary epithelial cells, selected for their ROS-responsive properties. MLE-12 membrane-coated berberine nanomicelles achieve targeted pulmonary delivery, demonstrating superior therapeutic outcomes compared to free berberine. This system significantly downregulates inflammatory mediators and ameliorates LPS-induced lung injury in mice (Table 2).181

Table 2 Nano-TCM Active Ingredient Co-Delivery Systems for Inflammatory Lung Disease

Inflammatory Bowel Disease (IBD)

Galactose- or folate-modified nanocarriers significantly improve colitis treatment by enhancing resveratrol’s intestinal absorption and transcellular transport.109,110 Similarly, milk-derived exosomes effectively protect resveratrol from degradation in the harsh gastrointestinal environment while maintaining its bioactivity, leading to notable colitis alleviation in rat models.182 The SF nanoparticle-resveratrol system demonstrates therapeutic efficacy against colitis comparable to dexamethasone.183

ROS-mediated oxidative stress plays a pivotal role in IBD pathogenesis, making ROS targeting a promising therapeutic strategy. The pH/ROS dual-responsive PEG-chitosan nanomicellar system encapsulating quercetin shows minimal drug release under physiological conditions but achieves nearly complete quercetin release in inflamed intestinal regions of IBD mice. This targeted release profile results in significant suppression of pro-inflammatory mediators including TNF-α, IL-6, and iNOS.184

The CXCL12-CXCR4 axis represents another important target for IBD therapy, as CXCL12 is markedly upregulated in inflamed tissues. Researchers have developed CXCR4-rich cell membrane vesicles that naturally home to inflammatory intestinal regions. These biomimetic vesicles not only facilitate targeted delivery of curcumin but also help evade immune clearance and prolong circulation time due to their native membrane composition (Table 3).185

Table 3 Nano-TCM Active Ingredient Co-Delivery Systems for IBD

In both dextran sulfate sodium (DSS)-induced ulcerative colitis and LPS-stimulated macrophage inflammation models, berberine-loaded PLGA nanoparticles (BPL-NPs) exhibit superior water solubility and bioactivity compared to free berberine. The BPL-NPs effectively reduce intestinal epithelial cell apoptosis and restore gut barrier function through selective modulation of the IL-6/IL-6R signaling axis.186

Rheumatoid Arthritis (RA) and Osteoarthritis (OA)

The nano-resveratrol co-delivery system demonstrates excellent transdermal penetration capability, efficiently delivering resveratrol into joint cavities to alleviate RA symptoms.187 When administered via intra-articular injection, this system significantly reduces TNF-α levels in knee joints, providing therapeutic benefits in rat OA models.188

Cadmium telluride semiconductor nanoparticles have been developed to enhance quercetin delivery, showing remarkable efficacy in promoting cartilage regeneration in OA rats through dual mechanisms of augmenting antioxidant enzyme activity and suppressing inflammatory mediator expression.189

Another promising approach involves the exosome-curcumin system, which targets the regulatory gene hsa-miR-126-3p in chondrocyte metabolism. In IL-1β-induced OA mice, this system effectively reduces chondrocyte catabolism and inflammation by upregulating hsa-miR-126-3p expression and subsequently inhibiting ERK1/2, PI3K/AKT, and p38 phosphorylation.190 Combination therapy using PLGA nanoparticles co-loaded with curcumin and meloxicam demonstrates superior anti-inflammatory effects compared to single-drug treatments in joint inflammation models.191

The xanthan gum-silver nanoparticle-bergenin system represents an innovative low-dose therapeutic approach, exhibiting enhanced immunomodulatory and antioxidant properties compared to free bergenin.64 This formulation significantly ameliorates OA symptoms in rats through multi-target inhibition of ROS, pro-inflammatory cytokines (IL-1β, IL-6, TNF-α), and Toll-like receptors (TLR-2, TLR-4).

Similarly, the nanocarrier-CK system effectively mitigates cartilage defects in OA mice by modulating gene expression patterns – downregulating cartilage degradation, inflammation, and lipogenesis markers while upregulating PPAR expression (Table 4).192

Table 4 Nano-TCM Active Ingredient Co-Delivery Systems for RA and OA

Conclusion and Future Prospects

This review systematically summarizes the anti-inflammatory mechanisms of key TCM active ingredients (resveratrol, quercetin, curcumin, bergenin, honokiol, ginsenosides, and berberine), which primarily involve modulation of SIRT1/SIRT3-mediated regulation of NF-κB, MAPK, PI3K/AKT, STAT3, and NRF2/HO-1 signaling pathways. We further elucidate the critical relationship between mitochondrial oxidative stress and inflammatory responses. While these compounds show significant therapeutic potential, their clinical translation has been limited by poor bioavailability, necessitating the development of nano-TCM delivery systems to enhance their pharmacokinetic profiles and therapeutic efficacy.

Current research on nanocarrier systems demonstrates considerable promise for pharmacological applications. These systems offer several advantages, including established safety profiles, flexible administration routes, and both passive and active targeting capabilities that significantly improve the bioavailability of TCM compounds. Among exogenous nanocarriers, lipid-based systems are particularly valuable for targeted delivery due to their amphiphilic nature, structural stability, biocompatibility, and ease of surface modification. Similarly, polymeric nanoparticles are widely utilized for their high drug encapsulation efficiency. However, potential nanotoxicity associated with certain carriers (eg, inorganic nanoparticles and nanoemulsions) requires careful evaluation. Studies have documented that inorganic nanoparticle accumulation may lead to hepatotoxicity,193 with slow hepatic metabolism and prolonged retention being major contributing factors.194 For instance, 28-day oral administration of silver nanoparticles in rats caused dose-dependent chronic liver injury, evidenced by altered alkaline phosphatase and cholesterol levels, along with inflammatory infiltration in hepatic tissues.195 Comparable findings show that magnetite iron oxide nanoparticles induce chronic pulmonary inflammation and granuloma formation in mice.196 At the molecular level, mesoporous silica and titanium dioxide nanoparticles activate inflammasome pathways,197,198 while repeated silver nanoparticle exposure leads to thymic atrophy, splenomegaly, and lymphocyte suppression.199

Compared to synthetic nanocarriers, biomimetic nanocarriers utilize endogenous components (eg, erythrocytes, leukocytes, macrophages) through surface functionalization to achieve “self-camouflage”, exhibiting superior properties such as reduced immunogenicity, enhanced biocompatibility, prolonged circulation time, and targeted accumulation at disease sites.19 However, the limited availability of natural source materials poses substantial challenges for large-scale production. Artificial exosomes have emerged as a promising alternative to natural exosomes and warrant further exploration.200 Current technological constraints continue to impede the translation of nanocarrier systems from preclinical research to industrial-scale manufacturing.201 There is an urgent need for comprehensive characterization of nanocarrier properties, including physicochemical parameters, enzymatic stability, morphological characteristics, and storage stability. Parallel research efforts should focus on developing nanocarriers with optimal safety profiles, structural stability, biocompatibility, controlled release kinetics, and high drug-loading capacity. To enhance targeting precision, various surface modification strategies have been investigated, employing ligands, macromolecules (monosaccharides, polysaccharides, peptides, folic acid, antibodies), surfactants, synthetic/natural polymers, and metallic components for nanocarrier functionalization.202

In inflammation-related diseases, numerous studies have reported nanocarrier systems delivering resveratrol, quercetin, and curcumin. However, research on nanocarriers incorporating honokiol, bergenin, and ginsenosides, and berberine remains limited despite their potent anti-inflammatory and antioxidant properties. Notably, clinical studies evaluating nano-formulated TCM active ingredient delivery systems are still insufficient. Importantly, nanocarriers co-delivering multiple active ingredients and/or therapeutic agents demonstrate superior efficacy compared to single-component formulations. This finding suggests a novel therapeutic strategy: combining nano-formulated TCM systems with conventional medications may achieve enhanced therapeutic outcomes at reduced and safer doses. For functionalized nanocarriers, a key challenge remains the identification of specific receptors and cytokines at lesion sites to enable precise targeting.

Data Sharing Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work. Chaoqun Sun, Ju Chen, and Shuyou Bai shared the first authorship.

Funding

This work was supported by the Youth Science Fund Project of National Natural Science Foundation of China (81600698), Science and Technology Planning Project of Guangdong Province (2023A1414020048), Medical Science and Technology Research Fund Project of Guangdong Province (B2024006), Medical Science and Technology Research Fund Project of Guangdong Province (B2024110).

Disclosure

The authors declare no conflicts of interest.

References

1. Chen L, Deng H, Cui H, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9(6):7204–7218. doi:10.18632/oncotarget.23208

2. Thuan DTB, Zayed H, Eid AH, et al. A potential link between oxidative stress and endothelial-to-mesenchymal transition in systemic sclerosis. Front Immunol. 2018;9:1985. doi:10.3389/fimmu.2018.01985

3. Li C, Lu Y, Du S, et al. Dioscin exerts protective effects against crystalline silica-induced pulmonary fibrosis in mice. Theranostics. 2017;7(17):4255–4275. doi:10.7150/thno.20270

4. He Y, Yue Y, Zheng X, Zhang K, Chen S, Du Z. Curcumin, inflammation, and chronic diseases: how are they linked? Molecules. 2015;20(5):9183–9213. doi:10.3390/molecules20059183

5. Cichoz-Lach H, Celinski K. Disadvantageous effects of nonsteroidal anti-inflammatory drugs on the alimentary tract. Ann Univ Mariae Curie Sklodowska Med. 2002;57(2):403–408.

6. Thushara RM, Hemshekhar M, Kemparaju K, Rangappa KS, Devaraja S, Girish KS. Therapeutic drug-induced platelet apoptosis: an overlooked issue in pharmacotoxicology. Arch Toxicol. 2014;88(2):185–198. doi:10.1007/s00204-013-1185-3

7. Mo C, Zhao J, Liang J, Wang H, Chen Y, Huang G. Exosomes: a novel insight into traditional Chinese medicine. Front Pharmacol. 2022;13:844782. doi:10.3389/fphar.2022.844782

8. Huang K, Zhang P, Zhang Z, et al. Traditional Chinese Medicine (TCM) in the treatment of COVID-19 and other viral infections: efficacies and mechanisms. Pharmacol Ther. 2021;225:107843. doi:10.1016/j.pharmthera.2021.107843

9. Wei Z, Chen J, Zuo F, et al. Traditional Chinese Medicine has great potential as candidate drugs for lung cancer: a review. J Ethnopharmacol. 2023;300:115748. doi:10.1016/j.jep.2022.115748

10. Zhou J, Yue J, Yao Y, et al. Dihydromyricetin protects intestinal barrier integrity by promoting IL-22 expression in ILC3s through the AMPK/SIRT3/STAT3 signaling pathway. Nutrients. 2023;15(2).

11. Cottart CH, Nivet-Antoine V, Laguillier-Morizot C, Beaudeux JL. Resveratrol bioavailability and toxicity in humans. Mol Nutr Food Res. 2010;54(1):7–16. doi:10.1002/mnfr.200900437

12. Dong X, Fu J, Yin X, et al. Emodin: a review of its pharmacology, toxicity and pharmacokinetics. Phytother Res. 2016;30(8):1207–1218. doi:10.1002/ptr.5631

13. Zhang Y, Qu Q, Zhao XM, et al. Medicinal evolution and modern research progress on Mume Fructus. Zhongguo Zhong Yao Za Zhi. 2023;48(14):3753–3764. doi:10.19540/j.cnki.cjcmm.20230331.302

14. Kim JH, Kwong EM, Chung VC, Lee JC, Wong T, Goggins WB. Acute adverse events from over-the-counter Chinese herbal medicines: a population-based survey of Hong Kong Chinese. BMC Complement Altern Med. 2013;13:336. doi:10.1186/1472-6882-13-336

15. Qiao Q, Liu X, Yang T, et al. Nanomedicine for acute respiratory distress syndrome: the latest application, targeting strategy, and rational design. Acta Pharm Sin B. 2021;11(10):3060–3091. doi:10.1016/j.apsb.2021.04.023

16. Quispe C, Cruz-Martins N, Manca ML, et al. Nano-derived therapeutic formulations with curcumin in inflammation-related diseases. Oxid Med Cell Longev. 2021;2021:3149223. doi:10.1155/2021/3149223

17. Liu Y, Feng N. Nanocarriers for the delivery of active ingredients and fractions extracted from natural products used in traditional Chinese medicine (TCM). Adv Colloid Interface Sci. 2015;221:60–76. doi:10.1016/j.cis.2015.04.006

18. Zhang Y, Huang Y, Li S. Polymeric micelles: nanocarriers for cancer-targeted drug delivery. AAPS Pharm Sci Tech. 2014;15(4):862–871. doi:10.1208/s12249-014-0113-z

19. Liu Q, Zou J, Chen Z, He W, Wu W. Current research trends of nanomedicines. Acta Pharm Sin B. 2023;13(11):4391–4416. doi:10.1016/j.apsb.2023.05.018

20. Galiniak S, Aebisher D, Bartusik-Aebisher D. Health benefits of resveratrol administration. Acta Biochim Pol. 2019;66(1):13–21. doi:10.18388/abp.2018_2749

21. Gal R, Deres L, Toth K, Halmosi R, Habon T. The effect of resveratrol on the cardiovascular system from molecular mechanisms to clinical results. Int J Mol Sci. 2021;22(18):10152. doi:10.3390/ijms221810152

22. Wang B, Sun J, Li X, et al. Resveratrol prevents suppression of regulatory T-cell production, oxidative stress, and inflammation of mice prone or resistant to high-fat diet-induced obesity. Nutr Res. 2013;33(11):971–981. doi:10.1016/j.nutres.2013.07.016

23. Andre DM, Calixto MC, Sollon C, et al. High-fat diet-induced obesity impairs insulin signaling in lungs of allergen-challenged mice: improvement by resveratrol. Sci Rep. 2017;7(1):17296. doi:10.1038/s41598-017-17558-w

24. Jeon BT, Jeong EA, Shin HJ, et al. Resveratrol attenuates obesity-associated peripheral and central inflammation and improves memory deficit in mice fed a high-fat diet. Diabetes. 2012;61(6):1444–1454. doi:10.2337/db11-1498

25. Singh UP, Singh NP, Singh B, et al. Resveratrol (trans-3,5,4’-trihydroxystilbene) induces silent mating type information regulation-1 and down-regulates nuclear transcription factor-kappaB activation to abrogate dextran sulfate sodium-induced colitis. J Pharmacol Exp Ther. 2010;332(3):829–839. doi:10.1124/jpet.109.160838

26. Elmali N, Esenkaya I, Harma A, Ertem K, Turkoz Y, Mizrak B. Effect of resveratrol in experimental osteoarthritis in rabbits. Inflamm Res. 2005;54(4):158–162. doi:10.1007/s00011-004-1341-6

27. Mattison JA, Wang M, Bernier M, et al. Resveratrol prevents high fat/sucrose diet-induced central arterial wall inflammation and stiffening in nonhuman primates. Cell Metab. 2014;20(1):183–190. doi:10.1016/j.cmet.2014.04.018

28. Li C, Wang Z, Lei H, Zhang D. Recent progress in nanotechnology-based drug carriers for resveratrol delivery. Drug Deliv. 2023;30(1):2174206. doi:10.1080/10717544.2023.2174206

29. Ebrahimpour S, Zakeri M, Esmaeili A. Crosstalk between obesity, diabetes, and Alzheimer’s disease: introducing quercetin as an effective triple herbal medicine. Ageing Res Rev. 2020;62:101095. doi:10.1016/j.arr.2020.101095

30. Chuang CC, Martinez K, Xie G, et al. Quercetin is equally or more effective than resveratrol in attenuating tumor necrosis factor-alpha-mediated inflammation and insulin resistance in primary human adipocytes. Am J Clin Nutr. 2010;92(6):1511–1521. doi:10.3945/ajcn.2010.29807

31. Lu H, Wu L, Liu L, et al. Quercetin ameliorates kidney injury and fibrosis by modulating M1/M2 macrophage polarization. Biochem Pharmacol. 2018;154:203–212. doi:10.1016/j.bcp.2018.05.007

32. Li X, Jin Q, Yao Q, et al. The flavonoid quercetin ameliorates liver inflammation and fibrosis by regulating hepatic macrophages activation and polarization in mice. Front Pharmacol. 2018;9:72. doi:10.3389/fphar.2018.00072

33. Feng K, Chen Z, Pengcheng L, Zhang S, Wang X. Quercetin attenuates oxidative stress-induced apoptosis via SIRT1/AMPK-mediated inhibition of ER stress in rat chondrocytes and prevents the progression of osteoarthritis in a rat model. J Cell Physiol. 2019;234(10):18192–18205. doi:10.1002/jcp.28452

34. Hu Y, Gui Z, Zhou Y, Xia L, Lin K, Xu Y. Quercetin alleviates rat osteoarthritis by inhibiting inflammation and apoptosis of chondrocytes, modulating synovial macrophages polarization to M2 macrophages. Free Radic Biol Med. 2019;145:146–160. doi:10.1016/j.freeradbiomed.2019.09.024

35. Shishehbor F, Behroo L, Ghafouriyan Broujerdnia M, Namjoyan F, Latifi SM. Quercetin effectively quells peanut-induced anaphylactic reactions in the peanut sensitized rats. Iran J Allergy Asthma Immunol. 2010;9(1):27–34.

36. Moon JH, Nakata R, Oshima S, Inakuma T, Terao J. Accumulation of quercetin conjugates in blood plasma after the short-term ingestion of onion by women. Am J Physiol Regul Integr Comp Physiol. 2000;279(2):R461–467. doi:10.1152/ajpregu.2000.279.2.R461

37. Mullen W, Edwards CA, Crozier A. Absorption, excretion and metabolite profiling of methyl-, glucuronyl-, glucosyl- and sulpho-conjugates of quercetin in human plasma and urine after ingestion of onions. Br J Nutr. 2006;96(1):107–116. doi:10.1079/BJN20061809

38. Slika L, Patra D. Traditional uses, therapeutic effects and recent advances of curcumin: a mini-review. Mini Rev Med Chem. 2020;20(12):1072–1082. doi:10.2174/1389557520666200414161316

39. Kao NJ, Hu JY, Wu CS, Kong ZL. Curcumin represses the activity of inhibitor-kappaB kinase in dextran sulfate sodium-induced colitis by S-nitrosylation. Int Immunopharmacol. 2016;38:1–7. doi:10.1016/j.intimp.2016.05.015

40. Mohammadian Haftcheshmeh S, Khosrojerdi A, Aliabadi A, Lotfi S, Mohammadi A, Momtazi-Borojeni AA. Immunomodulatory effects of curcumin in rheumatoid arthritis: evidence from molecular mechanisms to clinical outcomes. Rev Physiol Biochem Pharmacol. 2021;179:1–29. doi:10.1007/112_2020_54

41. Chen H, Tan H, Wan J, et al. PPAR-gamma signaling in nonalcoholic fatty liver disease: pathogenesis and therapeutic targets. Pharmacol Ther. 2023;245:108391. doi:10.1016/j.pharmthera.2023.108391

42. Li HY, Yang M, Li Z, Meng Z. Curcumin inhibits angiotensin II-induced inflammation and proliferation of rat vascular smooth muscle cells by elevating PPAR-gamma activity and reducing oxidative stress. Int J Mol Med. 2017;39(5):1307–1316. doi:10.3892/ijmm.2017.2924

43. Zhang Y, Liu Z, Wu J, et al. New MD2 inhibitors derived from curcumin with improved anti-inflammatory activity. Eur J Med Chem. 2018;148:291–305. doi:10.1016/j.ejmech.2018.02.008

44. Song Z, Revelo X, Shao W, et al. Dietary curcumin intervention targets mouse white adipose tissue inflammation and brown adipose tissue UCP1 expression. Obesity. 2018;26(3):547–558. doi:10.1002/oby.22110

45. Xu XY, Meng X, Li S, Gan RY, Li Y, Li HB. Bioactivity, health benefits, and related molecular mechanisms of curcumin: current progress, challenges, and perspectives. Nutrients. 2018;10(10):1553. doi:10.3390/nu10101553

46. Hewlings SJ, Kalman DS. Curcumin: a review of its effects on human health. Foods. 2017;6(10):92. doi:10.3390/foods6100092

47. Rauf A, Olatunde A, Imran M, et al. Honokiol: a review of its pharmacological potential and therapeutic insights. Phytomedicine. 2021;90:153647. doi:10.1016/j.phymed.2021.153647

48. Zhang Y, Liu Y, Hou M, et al. Reprogramming of mitochondrial respiratory chain complex by targeting SIRT3-COX4I2 axis attenuates osteoarthritis progression. Adv Sci. 2023;10(10):e2206144. doi:10.1002/advs.202206144

49. Jin L, Galonek H, Israelian K, et al. Biochemical characterization, localization, and tissue distribution of the longer form of mouse SIRT3. Protein Sci. 2009;18(3):514–525. doi:10.1002/pro.50

50. Chen X, Zhang M, Zhou F, et al. SIRT3 activator honokiol inhibits Th17 cell differentiation and alleviates colitis. Inflamm Bowel Dis. 2023;29(12):1929–1940. doi:10.1093/ibd/izad099

51. Wang L, Wang J. Honokiol ameliorates DSS-induced mouse colitis by inhibiting inflammation and oxidative stress and improving the intestinal barrier. Oxid Med Cell Longev. 2022;2022:1755608. doi:10.1155/2022/1755608

52. Liu J, Tang M, Li T, et al. Honokiol ameliorates post-myocardial infarction heart failure through Ucp3-mediated reactive oxygen species inhibition. Front Pharmacol. 2022;13:811682. doi:10.3389/fphar.2022.811682

53. Chen L, Li W, Qi D, Lu L, Zhang Z, Wang D. Honokiol protects pulmonary microvascular endothelial barrier against lipopolysaccharide-induced ARDS partially via the Sirt3/AMPK signaling axis. Life Sci. 2018;210:86–95. doi:10.1016/j.lfs.2018.08.064

54. Li F, Ye C, Wang X, Li X, Wang X. Honokiol ameliorates cigarette smoke-induced damage of airway epithelial cells via the SIRT3/SOD2 signalling pathway. J Cell Mol Med. 2023;27(24):4009–4020. doi:10.1111/jcmm.17981

55. Khatoon F, Ali S, Kumar V, et al. Pharmacological features, health benefits and clinical implications of honokiol. J Biomol Struct Dyn. 2023;41(15):7511–7533. doi:10.1080/07391102.2022.2120541

56. Sheng YL, Xu JH, Shi CH, et al. UPLC-MS/MS-ESI assay for simultaneous determination of magnolol and honokiol in rat plasma: application to pharmacokinetic study after administration emulsion of the isomer. J Ethnopharmacol. 2014;155(3):1568–1574. doi:10.1016/j.jep.2014.07.052

57. Salimo ZM, Yakubu MN, da Silva EL, et al. Chemistry and pharmacology of Bergenin or its derivatives: a promising molecule. Biomolecules. 2023;13(3):403. doi:10.3390/biom13030403

58. Huang D, Sun C, Chen M, et al. Bergenin ameliorates airway inflammation and remodeling in asthma by activating SIRT1 in macrophages to regulate the NF-kappaB pathway. Front Pharmacol. 2022;13:994878. doi:10.3389/fphar.2022.994878

59. Nazir N, Koul S, Qurishi MA, et al. Immunomodulatory effect of bergenin and norbergenin against adjuvant-induced arthritis–a flow cytometric study. J Ethnopharmacol. 2007;112(2):401–405. doi:10.1016/j.jep.2007.02.023

60. Wang K, Li YF, Lv Q, Li XM, Dai Y, Wei ZF. Bergenin, acting as an agonist of PPARgamma, ameliorates experimental colitis in mice through improving expression of SIRT1, and therefore inhibiting NF-kappaB-mediated macrophage activation. Front Pharmacol. 2017;8:981. doi:10.3389/fphar.2017.00981

61. Singla RK, Dhonchak K, Sodhi RK, et al. Bergenin ameliorates cognitive deficits and neuropathological alterations in sodium azide-induced experimental dementia. Front Pharmacol. 2022;13:994018. doi:10.3389/fphar.2022.994018

62. Ji Y, Wang D, Zhang B, Lu H. Bergenin ameliorates MPTP-induced Parkinson’s disease by activating PI3K/Akt signaling pathway. J Alzheimers Dis. 2019;72(3):823–833. doi:10.3233/JAD-190870

63. Zhou D, Qin X, Zhang ZR, Huang Y. Physicochemical properties of bergenin. Pharmazie. 2008;63(5):366–371.

64. Rao K, Roome T, Aziz S, et al. Bergenin loaded gum xanthan stabilized silver nanoparticles suppress synovial inflammation through modulation of the immune response and oxidative stress in adjuvant induced arthritic rats. J Mater Chem B. 2018;6(27):4486–4501. doi:10.1039/C8TB00672E

65. Qin X, Yang Y, Fan TT, Gong T, Zhang XN, Huang Y. Preparation, characterization and in vivo evaluation of bergenin-phospholipid complex. Acta Pharmacol Sin. 2010;31(1):127–136. doi:10.1038/aps.2009.171

66. Hu QR, Hong H, Zhang ZH, et al. Methods on improvements of the poor oral bioavailability of ginsenosides: pre-processing, structural modification, drug combination, and micro- or nano- delivery system. J Ginseng Res. 2023;47(6):694–705. doi:10.1016/j.jgr.2023.07.005

67. Paik S, Song GY, Jo EK. Ginsenosides for therapeutically targeting inflammation through modulation of oxidative stress. Int Immunopharmacol. 2023;121:110461. doi:10.1016/j.intimp.2023.110461

68. Kanai K, Kondo E. Antibacterial and cytotoxic aspects of long-chain fatty acids as cell surface events: selected topics. Jpn J Med Sci Biol. 1979;32(3):135–174. doi:10.7883/yoken1952.32.135

69. Kim DH, Chung JH, Yoon JS, et al. Ginsenoside Rd inhibits the expressions of iNOS and COX-2 by suppressing NF-kappaB in LPS-stimulated RAW264.7 cells and mouse liver. J Ginseng Res. 2013;37(1):54–63. doi:10.5142/jgr.2013.37.54

70. Xu K, Hu B, Ding X, Zhan Z. Alleviation of D-gal-induced senile liver injury by Rg3, a signature component of red ginseng. Aging. 2023;15(14):6749–6756. doi:10.18632/aging.204819

71. Joh EH, Lee IA, Jung IH, Kim DH. Ginsenoside Rb1 and its metabolite compound K inhibit IRAK-1 activation–the key step of inflammation. Biochem Pharmacol. 2011;82(3):278–286. doi:10.1016/j.bcp.2011.05.003

72. Lee SY, Jeong JJ, Eun SH, Kim DH. Anti-inflammatory effects of ginsenoside Rg1 and its metabolites ginsenoside Rh1 and 20(S)-protopanaxatriol in mice with TNBS-induced colitis. Eur J Pharmacol. 2015;762:333–343. doi:10.1016/j.ejphar.2015.06.011

73. Kim DH, Kim DW, Jung BH, et al. Ginsenoside Rb2 suppresses the glutamate-mediated oxidative stress and neuronal cell death in HT22 cells. J Ginseng Res. 2019;43(2):326–334. doi:10.1016/j.jgr.2018.12.002

74. Xue Y, Fu W, Liu Y, et al. Ginsenoside Rb2 alleviates myocardial ischemia/reperfusion injury in rats through SIRT1 activation. J Food Sci. 2020;85(11):4039–4049. doi:10.1111/1750-3841.15505

75. Xue Y, Fu W, Yu P, et al. Ginsenoside Rc alleviates myocardial ischemia-reperfusion injury by reducing mitochondrial oxidative stress and apoptosis: role of SIRT1 activation. J Agric Food Chem. 2023;71(3):1547–1561. doi:10.1021/acs.jafc.2c06926

76. Wang QL, Yang L, Peng Y, et al. Ginsenoside Rg1 regulates SIRT1 to ameliorate sepsis-induced lung inflammation and injury via inhibiting endoplasmic reticulum stress and inflammation. Mediators Inflamm. 2019;2019:6453296. doi:10.1155/2019/6453296

77. Ma CH, Chou WC, Wu CH, et al. Ginsenoside Rg3 attenuates TNF-alpha-induced damage in chondrocytes through regulating SIRT1-mediated anti-apoptotic and anti-inflammatory mechanisms. Antioxidants. 2021;10(12). doi:10.3390/antiox10121972

78. Lee YY, Park JS, Lee EJ, et al. Anti-inflammatory mechanism of ginseng saponin metabolite Rh3 in lipopolysaccharide-stimulated microglia: critical role of 5’-adenosine monophosphate-activated protein kinase signaling pathway. J Agric Food Chem. 2015;63(13):3472–3480. doi:10.1021/jf506110y

79. Wu K, Shieh JS, Qin L, Guo JJ. Mitochondrial mechanisms in the pathogenesis of chronic inflammatory musculoskeletal disorders. Cell Biosci. 2024;14(1):76. doi:10.1186/s13578-024-01259-9

80. Xing W, Yang L, Peng Y, et al. Ginsenoside Rg3 attenuates sepsis-induced injury and mitochondrial dysfunction in liver via AMPK-mediated autophagy flux. Biosci Rep. 2017;37(4). doi:10.1042/BSR20170934

81. Lee SJ, Bae JH, Lee H, et al. Ginsenoside Rg3 upregulates myotube formation and mitochondrial function, thereby protecting myotube atrophy induced by tumor necrosis factor-alpha. J Ethnopharmacol. 2019;242:112054. doi:10.1016/j.jep.2019.112054

82. Paik S, Choe JH, Choi GE, et al. Rg6, a rare ginsenoside, inhibits systemic inflammation through the induction of interleukin-10 and microRNA-146a. Sci Rep. 2019;9(1):4342. doi:10.1038/s41598-019-40690-8

83. Jin Y, Nguyen TLL, Myung CS, Heo KS. Ginsenoside Rh1 protects human endothelial cells against lipopolysaccharide-induced inflammatory injury through inhibiting TLR2/4-mediated STAT3, NF-kappaB, and ER stress signaling pathways. Life Sci. 2022;309:120973. doi:10.1016/j.lfs.2022.120973

84. Miao L, Yang Y, Li Z, Fang Z, Zhang Y, Han CC. Ginsenoside Rb2: a review of pharmacokinetics and pharmacological effects. J Ginseng Res. 2022;46(2):206–213. doi:10.1016/j.jgr.2021.11.007

85. Lu Q, Fu Y, Li H. Berberine and its derivatives represent as the promising therapeutic agents for inflammatory disorders. Pharmacol Rep. 2022;74(2):297–309. doi:10.1007/s43440-021-00348-7

86. Maskey AR, Kopulos D, Kwan M, et al. Berberine inhibits the inflammatory response induced by Staphylococcus aureus isolated from atopic eczema patients via the TNF-alpha/inflammation/RAGE pathways. Cells. 2024;13(19):1639. doi:10.3390/cells13191639

87. Jeong HW, Hsu KC, Lee JW, et al. Berberine suppresses proinflammatory responses through AMPK activation in macrophages. Am J Physiol Endocrinol Metab. 2009;296(4):E955–964. doi:10.1152/ajpendo.90599.2008

88. Li Z, Geng YN, Jiang JD, Kong WJ. Antioxidant and anti-inflammatory activities of berberine in the treatment of diabetes mellitus. Evid Based Complement Alternat Med. 2014;2014:289264. doi:10.1155/2014/289264

89. Yang N, Wang J, Liu C, et al. Berberine and limonin suppress IgE production by human B cells and peripheral blood mononuclear cells from food-allergic patients. Ann Allergy Asthma Immunol. 2014;113(5):556–564e554. doi:10.1016/j.anai.2014.07.021

90. Srivastava K, Cao M, Fidan O, et al. Berberine-containing natural-medicine with boiled peanut-OIT induces sustained peanut-tolerance associated with distinct microbiota signature. Front Immunol. 2023;14:1174907. doi:10.3389/fimmu.2023.1174907

91. Liu CS, Zheng YR, Zhang YF, Long XY. Research progress on berberine with a special focus on its oral bioavailability. Fitoterapia. 2016;109:274–282. doi:10.1016/j.fitote.2016.02.001

92. Spinozzi S, Colliva C, Camborata C, et al. Berberine and its metabolites: relationship between physicochemical properties and plasma levels after administration to human subjects. J Nat Prod. 2014;77(4):766–772. doi:10.1021/np400607k

93. Zu Y, Overby H, Ren G, Fan Z, Zhao L, Wang S. Resveratrol liposomes and lipid nanocarriers: comparison of characteristics and inducing browning of white adipocytes. Colloids Surf B Biointerfaces. 2018;164:414–423. doi:10.1016/j.colsurfb.2017.12.044

94. Sun L, Hu Y, Mishra A, et al. Protective role of poly(lactic-co-glycolic) acid nanoparticle loaded with resveratrol against isoproterenol-induced myocardial infarction. Biofactors. 2020;46(3):421–431. doi:10.1002/biof.1611

95. Wang L, Xu X, Zhang Y, et al. Encapsulation of curcumin within poly(amidoamine) dendrimers for delivery to cancer cells. J Mater Sci Mater Med. 2013;24(9):2137–2144. doi:10.1007/s10856-013-4969-3

96. Guo Y, Zhao Y, Wang T, et al. Honokiol nanoparticles stabilized by oligoethylene glycols codendrimer: in vitro and in vivo investigations. J Mater Chem B. 2017;5(4):697–706. doi:10.1039/C6TB02416E

97. Yang F, Zhou J, Hu X, et al. Preparation and evaluation of self-microemulsions for improved bioavailability of ginsenoside-Rh1 and Rh2. Drug Deliv Transl Res. 2017;7(5):731–737. doi:10.1007/s13346-017-0402-7

98. Ezeh CK, Dibua MEU. Anti-biofilm, drug delivery and cytotoxicity properties of dendrimers. ADMET DMPK. 2024;12(2):239–267. doi:10.5599/admet.1917

99. Wang X, Zhao X, Zhong Y, Shen J, An W. Biomimetic exosomes: a new generation of drug delivery system. Front Bioeng Biotechnol. 2022;10:865682. doi:10.3389/fbioe.2022.865682

100. Cai S, Shi CH, Zhang X, et al. Self-microemulsifying drug-delivery system for improved oral bioavailability of 20(S)-25-methoxyl-dammarane-3beta, 12beta, 20-triol: preparation and evaluation. Int J Nanomed. 2014;9:913–920. doi:10.2147/IJN.S56894

101. Cheng Y, Xu Z, Ma M, Xu T. Dendrimers as drug carriers: applications in different routes of drug administration. J Pharm Sci. 2008;97(1):123–143. doi:10.1002/jps.21079

102. Li S, Chen L, Wang G, et al. Anti-ICAM-1 antibody-modified nanostructured lipid carriers: a pulmonary vascular endothelium-targeted device for acute lung injury therapy. J Nanobiotechnology. 2018;16(1):105. doi:10.1186/s12951-018-0431-5

103. Jin F, Liu D, Yu H, et al. Sialic acid-functionalized PEG-PLGA microspheres loading mitochondrial-targeting-modified curcumin for acute lung injury therapy. Mol Pharm. 2019;16(1):71–85. doi:10.1021/acs.molpharmaceut.8b00861

104. Herber-Jonat S, Mittal R, Gsinn S, Bohnenkamp H, Guenzi E, Schulze A. Comparison of lung accumulation of cationic liposomes in normal rats and LPS-treated rats. Inflamm Res. 2011;60(3):245–253. doi:10.1007/s00011-010-0260-y

105. Kotta S, Mubarak Aldawsari H, Badr-Eldin SM, Alhakamy NA, Md S. Coconut oil-based resveratrol nanoemulsion: optimization using response surface methodology, stability assessment and pharmacokinetic evaluation. Food Chem. 2021;357:129721. doi:10.1016/j.foodchem.2021.129721

106. Yu Y, Lu Y, Bo R, et al. The preparation of gypenosides liposomes and its effects on the peritoneal macrophages function in vitro. Int J Pharm. 2014;460(1–2):248–254. doi:10.1016/j.ijpharm.2013.11.018

107. Zheng X, Sun K, Liu Y, et al. Resveratrol-loaded macrophage exosomes alleviate multiple sclerosis through targeting microglia. J Control Release. 2023;353:675–684. doi:10.1016/j.jconrel.2022.12.026

108. Araujo EV, Carneiro SV, Neto DMA, et al. Advances in surface design and biomedical applications of magnetic nanoparticles. Adv Colloid Interface Sci. 2024;328:103166. doi:10.1016/j.cis.2024.103166

109. Naserifar M, Hosseinzadeh H, Abnous K, et al. Oral delivery of folate-targeted resveratrol-loaded nanoparticles for inflammatory bowel disease therapy in rats. Life Sci. 2020;262:118555. doi:10.1016/j.lfs.2020.118555

110. Siu FY, Ye S, Lin H, Li S. Galactosylated PLGA nanoparticles for the oral delivery of resveratrol: enhanced bioavailability and in vitro anti-inflammatory activity. Int J Nanomed. 2018;13:4133–4144. doi:10.2147/IJN.S164235

111. Katila N, Duwa R, Bhurtel S, et al. Enhancement of blood-brain barrier penetration and the neuroprotective effect of resveratrol. J Control Release. 2022;346:1–19. doi:10.1016/j.jconrel.2022.04.003

112. Jaynes JM, Sable R, Ronzetti M, et al. Mannose receptor (CD206) activation in tumor-associated macrophages enhances adaptive and innate antitumor immune responses. Sci Transl Med. 2020;12(530). doi:10.1126/scitranslmed.aax6337

113. Zheng X, Wu F, Hong Y, Shen L, Lin X, Feng Y. Developments in taste-masking techniques for Traditional Chinese Medicines. Pharmaceutics. 2018;10(3):157. doi:10.3390/pharmaceutics10030157

114. Cui Y, Zhou Q, Jin M, et al. Research progress on pharmacological effects and bioavailability of berberine. Naunyn Schmiedebergs Arch Pharmacol. 2024;397(11):8485–8514. doi:10.1007/s00210-024-03199-0

115. Cao X, Deng WW, Fu M, et al. In vitro release and in vitro-in vivo correlation for silybin meglumine incorporated into hollow-type mesoporous silica nanoparticles. Int J Nanomed. 2012;7:753–762. doi:10.2147/IJN.S28348

116. Wang X, Wan W, Lu J, Liu P. Inhalable FN-binding liposomes or liposome-exosome hybrid bionic vesicles encapsulated microparticles for enhanced pulmonary fibrosis therapy. Int J Pharm. 2024;656:124096. doi:10.1016/j.ijpharm.2024.124096

117. Abri Aghdam M, Bagheri R, Mosafer J, et al. Recent advances on thermosensitive and pH-sensitive liposomes employed in controlled release. J Control Release. 2019;315:1–22. doi:10.1016/j.jconrel.2019.09.018

118. Zhang C, Gu C, Peng F, et al. Preparation and optimization of triptolide-loaded solid lipid nanoparticles for oral delivery with reduced gastric irritation. Molecules. 2013;18(11):13340–13356. doi:10.3390/molecules181113340

119. Barui S, Saha S, Mondal G, Haseena S, Chaudhuri A. Simultaneous delivery of doxorubicin and curcumin encapsulated in liposomes of pegylated RGDK-lipopeptide to tumor vasculature. Biomaterials. 2014;35(5):1643–1656. doi:10.1016/j.biomaterials.2013.10.074

120. Vaiss DP, Rodrigues JL, Yurgel VC, et al. Curcumin and quercetin co-encapsulated in nanoemulsions for nasal administration: a promising therapeutic and prophylactic treatment for viral respiratory infections. Eur J Pharm Sci. 2024;197:106766. doi:10.1016/j.ejps.2024.106766

121. Li Y, Guo C, Chen Q, et al. Improvement of pneumonia by curcumin-loaded bionanosystems based on platycodon grandiflorum polysaccharides via calming cytokine storm. Int J Biol Macromol. 2022;202:691–706. doi:10.1016/j.ijbiomac.2022.01.194

122. Tomou EM, Papakyriakopoulou P, Saitani EM, Valsami G, Pippa N, Skaltsa H. Recent advances in nanoformulations for Quercetin Delivery. Pharmaceutics. 2023;15(6):1656. doi:10.3390/pharmaceutics15061656

123. de Oliveira MTP, Coutinho DS, Guterres SS, et al. Resveratrol-loaded lipid-core nanocapsules modulate acute lung inflammation and oxidative imbalance induced by LPS in mice. Pharmaceutics. 2021;13(5):683. doi:10.3390/pharmaceutics13050683

124. Rashwan AK, Karim N, Xu Y, et al. An updated and comprehensive review on the potential health effects of curcumin-encapsulated micro/nanoparticles. Crit Rev Food Sci Nutr. 2023;63(29):9731–9751. doi:10.1080/10408398.2022.2070906

125. Scioli Montoto S, Muraca G, Ruiz ME. Solid lipid nanoparticles for drug delivery: pharmacological and biopharmaceutical aspects. Front Mol Biosci. 2020;7:587997. doi:10.3389/fmolb.2020.587997

126. Pink DL, Loruthai O, Ziolek RM, et al. On the structure of solid lipid nanoparticles. Small. 2019;15(45):e1903156. doi:10.1002/smll.201903156

127. Das S, Ng WK, Tan RB. Are nanostructured lipid carriers (NLCs) better than solid lipid nanoparticles (SLNs): development, characterizations and comparative evaluations of clotrimazole-loaded SLNs and NLCs? Eur J Pharm Sci. 2012;47(1):139–151. doi:10.1016/j.ejps.2012.05.010

128. Nnamani PO, Nwagwu C, Diovu EO, et al. Design and evaluation of nanostructured lipid carrier of Bergenin isolated from Pentaclethra macrophylla for anti-inflammatory effect on lipopolysaccharide-induced inflammatory responses in macrophages. Eur J Pharm Biopharm. 2024;200:114307. doi:10.1016/j.ejpb.2024.114307

129. Lucio M, Giannino N, Barreira S, et al. Nanostructured lipid carriers enriched hydrogels for skin topical administration of Quercetin and Omega-3 fatty acid. Pharmaceutics. 2023;15(8):2078. doi:10.3390/pharmaceutics15082078

130. Kim MH, Kim KT, Sohn SY, et al. Formulation and evaluation of nanostructured lipid carriers (NLCs) Of 20(S)-Protopanaxadiol (PPD) by Box-Behnken design. Int J Nanomed. 2019;14:8509–8520. doi:10.2147/IJN.S215835

131. Signorini S, Delledonne A, Pescina S, et al. A sterilizable platform based on crosslinked xanthan gum for controlled-release of polymeric micelles: ocular application for the delivery of neuroprotective compounds to the posterior eye segment. Int J Pharm. 2024;657:124141. doi:10.1016/j.ijpharm.2024.124141

132. Zu M, Ma Y, Cannup B, et al. Oral delivery of natural active small molecules by polymeric nanoparticles for the treatment of inflammatory bowel diseases. Adv Drug Deliv Rev. 2021;176:113887. doi:10.1016/j.addr.2021.113887

133. Menjoge AR, Kannan RM, Tomalia DA. Dendrimer-based drug and imaging conjugates: design considerations for nanomedical applications. Drug Discov Today. 2010;15(5–6):171–185. doi:10.1016/j.drudis.2010.01.009

134. Street CM, Zhu Z, Finel M, Court MH. Bisphenol-A glucuronidation in human liver and breast: identification of UDP-glucuronosyltransferases (UGTs) and influence of genetic polymorphisms. Xenobiotica. 2017;47(1):1–10. doi:10.3109/00498254.2016.1156784

135. Pissuwan D, Niidome T, Cortie MB. The forthcoming applications of gold nanoparticles in drug and gene delivery systems. J Control Release. 2011;149(1):65–71. doi:10.1016/j.jconrel.2009.12.006

136. Dai T, He W, Yao C, et al. Applications of inorganic nanoparticles in the diagnosis and therapy of atherosclerosis. Biomater Sci. 2020;8(14):3784–3799. doi:10.1039/D0BM00196A

137. Muller RH, Gohla S, Keck CM. State of the art of nanocrystals–special features, production, nanotoxicology aspects and intracellular delivery. Eur J Pharm Biopharm. 2011;78(1):1–9. doi:10.1016/j.ejpb.2011.01.007

138. Kemp MM, Linhardt RJ. Heparin-based nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2010;2(1):77–87. doi:10.1002/wnan.68

139. Yang X, Du H, Liu J, Zhai G. Advanced nanocarriers based on heparin and its derivatives for cancer management. Biomacromolecules. 2015;16(2):423–436. doi:10.1021/bm501532e

140. Wang Q, Duan Y, Jing H, et al. Inhibition of atherosclerosis progression by modular micelles. J Control Release. 2023;354:294–304. doi:10.1016/j.jconrel.2023.01.020

141. Liang X, Yang Y, Huang C, et al. cRGD-targeted heparin nanoparticles for effective dual drug treatment of cisplatin-resistant ovarian cancer. J Control Release. 2023;356:691–701. doi:10.1016/j.jconrel.2023.03.017

142. Li N, Zhao L, Geng X, et al. Stimulation by exosomes from hypoxia-preconditioned hair follicle mesenchymal stem cells facilitates mitophagy by inhibiting the PI3K/AKT/mTOR signaling pathway to alleviate ulcerative colitis. Theranostics. 2024;14(11):4278–4296. doi:10.7150/thno.96038

143. Li Y, Dong L, Liu Y, et al. Ultrasound and enzyme assisted preparation of novel lactoferrin-cereal phenolic acid conjugates: structural, physicochemical and functional properties. Food Chem. 2024;435:137572. doi:10.1016/j.foodchem.2023.137572

144. Iqbal H, Yang T, Li T, et al. Serum protein-based nanoparticles for cancer diagnosis and treatment. J Control Release. 2021;329:997–1022. doi:10.1016/j.jconrel.2020.10.030

145. Zhu R, Avsievich T, Popov A, Bykov A, Meglinski I. In vivo nano-biosensing element of red blood cell-mediated delivery. Biosens Bioelectron. 2021;175:112845. doi:10.1016/j.bios.2020.112845

146. Cinti C, Taranta M, Naldi I, Grimaldi S. Newly engineered magnetic erythrocytes for sustained and targeted delivery of anti-cancer therapeutic compounds. PLOS ONE. 2011;6(2):e17132. doi:10.1371/journal.pone.0017132

147. Hamidi M, Azimi K, Mohammadi-Samani S. Co-encapsulation of a drug with a protein in erythrocytes for improved drug loading and release: phenytoin and bovine serum albumin (BSA). J Pharm Pharm Sci. 2011;14(1):46–59. doi:10.18433/J37W2V

148. Han Y, Chu X, Cui L, et al. Neuronal mitochondria-targeted therapy for Alzheimer’s disease by systemic delivery of resveratrol using dual-modified novel biomimetic nanosystems. Drug Deliv. 2020;27(1):502–518. doi:10.1080/10717544.2020.1745328

149. Brenner JS, Pan DC, Myerson JW, et al. Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude. Nat Commun. 2018;9(1):2684. doi:10.1038/s41467-018-05079-7

150. Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release. 2020;327:70–87. doi:10.1016/j.jconrel.2020.07.043

151. Pan DC, Myerson JW, Brenner JS, et al. Nanoparticle properties modulate their attachment and effect on carrier red blood cells. Sci Rep. 2018;8(1):1615. doi:10.1038/s41598-018-19897-8

152. de la Harpe KM, Kondiah PPD, Choonara YE, Marimuthu T, du Toit LC, Pillay V. The hemocompatibility of nanoparticles: a review of cell-nanoparticle interactions and hemostasis. Cells. 2019;8(10):1209. doi:10.3390/cells8101209

153. Guo Q, Qian ZM. Macrophage based drug delivery: key challenges and strategies. Bioact Mater. 2024;38:55–72. doi:10.1016/j.bioactmat.2024.04.004

154. Scarpa E, Bailey JL, Janeczek AA, et al. Quantification of intracellular payload release from polymersome nanoparticles. Sci Rep. 2016;6:29460. doi:10.1038/srep29460

155. Sun P, Deng Q, Kang L, Sun Y, Ren J, Qu X. A smart nanoparticle-laden and remote-controlled self-destructive macrophage for enhanced chemo/chemodynamic synergistic therapy. ACS Nano. 2020;14(10):13894–13904. doi:10.1021/acsnano.0c06290

156. Stephan MT, Moon JJ, Um SH, Bershteyn A, Irvine DJ. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat Med. 2010;16(9):1035–1041. doi:10.1038/nm.2198

157. Mori T, Giovannelli L, Bilia AR, Margheri F. Exosomes: potential next-generation nanocarriers for the therapy of inflammatory diseases. Pharmaceutics. 2023;15(9):2276. doi:10.3390/pharmaceutics15092276

158. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016;371(1):48–61. doi:10.1016/j.canlet.2015.10.020

159. Seasons GM, Pellow C, Kuipers HF, Pike GB. Ultrasound and neuroinflammation: immune modulation via the heat shock response. Theranostics. 2024;14(8):3150–3177. doi:10.7150/thno.96270

160. Wang Z, Pan J, Yuan R, Chen M, Guo X, Zhou S. Shell-sheddable polymeric micelles alleviate oxidative stress and inflammation for enhanced ischemic stroke therapy. Nano Lett. 2023;23(14):6544–6552. doi:10.1021/acs.nanolett.3c01567

161. Li C, Wang N, Zheng G, Yang L. Oral administration of resveratrol-selenium-peptide nanocomposites alleviates Alzheimer’s disease-like pathogenesis by inhibiting Abeta aggregation and regulating gut microbiota. ACS Appl Mater Interfaces. 2021;13(39):46406–46420. doi:10.1021/acsami.1c14818

162. Abozaid OAR, Sallam MW, El-Sonbaty S, Aziza S, Emad B, Ahmed ESA. Resveratrol-Selenium nanoparticles alleviate neuroinflammation and neurotoxicity in a rat model of Alzheimer’s disease by regulating Sirt1/miRNA-134/GSK3beta expression. Biol Trace Elem Res. 2022;200(12):5104–5114. doi:10.1007/s12011-021-03073-7

163. Kaur S, Sharma K, Sharma A, et al. Fluvoxamine maleate alleviates amyloid-beta load and neuroinflammation in 5XFAD mice to ameliorate Alzheimer disease pathology. Front Immunol. 2024;15:1418422. doi:10.3389/fimmu.2024.1418422

164. Yang JT, Kuo YC, Chen IY, Rajesh R, Lou YI, Hsu JP. Protection against neurodegeneration in the hippocampus using Sialic Acid- and 5-HT-moduline-conjugated lipopolymer nanoparticles. ACS Biomater Sci Eng. 2019;5(3):1311–1320. doi:10.1021/acsbiomaterials.8b01334

165. Halevas E, Mavroidi B, Nday CM, et al. Modified magnetic core-shell mesoporous silica nano-formulations with encapsulated quercetin exhibit anti-amyloid and antioxidant activity. J Inorg Biochem. 2020;213:111271. doi:10.1016/j.jinorgbio.2020.111271

166. Qi Y, Guo L, Jiang Y, Shi Y, Sui H, Zhao L. Brain delivery of quercetin-loaded exosomes improved cognitive function in AD mice by inhibiting phosphorylated tau-mediated neurofibrillary tangles. Drug Deliv. 2020;27(1):745–755. doi:10.1080/10717544.2020.1762262

167. Wang Y, Luo J, Li SY. Nano-Curcumin simultaneously protects the blood-brain barrier and reduces M1 Microglial activation during cerebral ischemia-reperfusion injury. ACS Appl Mater Interfaces. 2019;11(4):3763–3770. doi:10.1021/acsami.8b20594

168. Qu C, Li QP, Su ZR, et al. Nano-Honokiol ameliorates the cognitive deficits in TgCRND8 mice of Alzheimer’s disease via inhibiting neuropathology and modulating gut microbiota. J Adv Res. 2022;35:231–243. doi:10.1016/j.jare.2021.03.012

169. Hsiao YP, Chen HT, Liang YC, et al. Development of nanosome-encapsulated Honokiol for intravenous therapy against experimental autoimmune encephalomyelitis. Int J Nanomed. 2020;15:17–29. doi:10.2147/IJN.S214349

170. Cheng Z, Zhang M, Ling C, et al. Neuroprotective effects of Ginsenosides against cerebral ischemia. Molecules. 2019;24(6):1102. doi:10.3390/molecules24061102

171. Shen J, Zhao Z, Shang W, et al. Ginsenoside Rg1 nanoparticle penetrating the blood-brain barrier to improve the cerebral function of diabetic rats complicated with cerebral infarction. Int J Nanomed. 2017;12:6477–6486. doi:10.2147/IJN.S139602

172. Liu T, Zhang M, Zhang J, Kang N, Zheng L, Ding Z. Targeted delivery of macrophage membrane biomimetic liposomes through intranasal administration for treatment of ischemic stroke. Int J Nanomed. 2024;19:6177–6199. doi:10.2147/IJN.S458656

173. Liu T, Wang Y, Zhang M, et al. The optimization design of macrophage membrane camouflaging liposomes for alleviating ischemic stroke injury through intranasal delivery. Int J Mol Sci. 2024;25(5).

174. de Oliveira MTP, De Sa coutinho D, Tenorio de Souza E, et al. Orally delivered resveratrol-loaded lipid-core nanocapsules ameliorate LPS-induced acute lung injury via the ERK and PI3K/Akt pathways. Int J Nanomed. 2019;14:5215–5228. doi:10.2147/IJN.S200666

175. Zheng X, Xing Y, Sun K, Jin H, Zhao W, Yu F. Combination therapy with resveratrol and celastrol using folic acid-functionalized exosomes enhances the therapeutic efficacy of sepsis. Adv Healthc Mater. 2023;12(29):e2301325. doi:10.1002/adhm.202301325

176. Xu X, Kwong CHT, Li J, Wei J, Wang R. “Zombie” macrophages for targeted drug delivery to treat acute pneumonia. ACS Appl Mater Interfaces. 2023;15(24):29012–29022. doi:10.1021/acsami.3c06025

177. Kim G, Lee Y, Ha J, Han S, Lee M. Engineering exosomes for pulmonary delivery of peptides and drugs to inflammatory lung cells by inhalation. J Control Release. 2021;330:684–695. doi:10.1016/j.jconrel.2020.12.053

178. Wang X, Wang Y, Tang T, et al. Curcumin-loaded RH60/F127 mixed micelles: characterization, biopharmaceutical characters and anti-inflammatory modulation of airway inflammation. Pharmaceutics. 2023;15(12):2710. doi:10.3390/pharmaceutics15122710

179. Lin H, Wang P, Zhang W, et al. Novel combined preparation and investigation of Bergenin-loaded albumin nanoparticles for the treatment of acute lung injury: in vitro and in vivo evaluations. Inflammation. 2022;45(1):428–444. doi:10.1007/s10753-021-01556-2

180. Park HH, Kim H, Lee HS, et al. PEGylated nanoparticle albumin-bound steroidal ginsenoside derivatives ameliorate SARS-CoV-2-mediated hyper-inflammatory responses. Biomaterials. 2021;273:120827. doi:10.1016/j.biomaterials.2021.120827

181. Jin C, Zhang Y, Chen L, et al. Lung epithelial cell membrane-camouflaged ROS-activatable Berberine nanoparticles for targeted treatment in acute lung injury. Int J Nanomed. 2025;20:6163–6183. doi:10.2147/IJN.S514611

182. Esfahani SK, Dehghani S, Hosseinzadeh H, et al. An exosomal approach for oral delivery of resveratrol: implications for inflammatory bowel disease treatment in rat model. Life Sci. 2024;346:122638. doi:10.1016/j.lfs.2024.122638

183. Lozano-Perez AA, Rodriguez-Nogales A, Ortiz-Cullera V, et al. Silk fibroin nanoparticles constitute a vector for controlled release of resveratrol in an experimental model of inflammatory bowel disease in rats. Int J Nanomed. 2014;9:4507–4520. doi:10.2147/IJN.S68526

184. Shen C, Zhao L, Du X, et al. Smart responsive quercetin-conjugated glycol chitosan prodrug micelles for treatment of inflammatory bowel diseases. Mol Pharm. 2021;18(3):1419–1430. doi:10.1021/acs.molpharmaceut.0c01245

185. Wang D, Jiang S, Zhang F, et al. Cell membrane vesicles with enriched CXCR4 display enhances their targeted delivery as drug carriers to inflammatory sites. Adv Sci. 2021;8(23):e2101562. doi:10.1002/advs.202101562

186. Liu C, Gong Q, Liu W, Zhao Y, Yan X, Yang T. Berberine-loaded PLGA nanoparticles alleviate ulcerative colitis by targeting IL-6/IL-6R axis. J Transl Med. 2024;22(1):963. doi:10.1186/s12967-024-05682-x

187. Diao N, Liu Y, Wang W, et al. Resveratrol nanocrystals based dissolving microneedles with highly efficient for rheumatoid arthritis. Drug Deliv Transl Res. 2025;15(1):203–215. doi:10.1007/s13346-024-01581-2

188. Kamel R, Abbas H, Shaffie NM. Development and evaluation of PLA-coated co-micellar nanosystem of Resveratrol for the intra-articular treatment of arthritis. Int J Pharm. 2019;569:118560. doi:10.1016/j.ijpharm.2019.118560

189. Jeyadevi R, Sivasudha T, Rameshkumar A, et al. Enhancement of anti arthritic effect of quercetin using thioglycolic acid-capped cadmium telluride quantum dots as nanocarrier in adjuvant induced arthritic Wistar rats. Colloids Surf B Biointerfaces. 2013;112:255–263. doi:10.1016/j.colsurfb.2013.07.065

190. Li S, Stockl S, Lukas C, et al. Curcumin-primed human BMSC-derived extracellular vesicles reverse IL-1beta-induced catabolic responses of OA chondrocytes by upregulating miR-126-3p. Stem Cell Res Ther. 2021;12(1):252. doi:10.1186/s13287-021-02317-6

191. Aslam B, Hussain A, Faisal MN, et al. Curcumin co-encapsulation potentiates anti-arthritic efficacy of meloxicam biodegradable nanoparticles in adjuvant-induced arthritis animal model. Biomedicines. 2023;11(10):2662. doi:10.3390/biomedicines11102662

192. Shin HH, Park J, Kim YJ, Kim D, Jin EJ, Ryu JH. Hydrophilic/hydrophobic Janus nanofibers containing compound K for cartilage regeneration. Int J Nanomed. 2024;19:1683–1697. doi:10.2147/IJN.S435156

193. Mohammadpour R, Dobrovolskaia MA, Cheney DL, Greish KF, Ghandehari H. Subchronic and chronic toxicity evaluation of inorganic nanoparticles for delivery applications. Adv Drug Deliv Rev. 2019;144:112–132. doi:10.1016/j.addr.2019.07.006

194. Sadauskas E, Danscher G, Stoltenberg M, Vogel U, Larsen A, Wallin H. Protracted elimination of gold nanoparticles from mouse liver. Nanomedicine. 2009;5(2):162–169. doi:10.1016/j.nano.2008.11.002

195. Kim YS, Kim JS, Cho HS, et al. Twenty-eight-day oral toxicity, genotoxicity, and gender-related tissue distribution of silver nanoparticles in Sprague-Dawley rats. Inhal Toxicol. 2008;20(6):575–583. doi:10.1080/08958370701874663

196. Park EJ, Kim H, Kim Y, Yi J, Choi K, Park K. Inflammatory responses may be induced by a single intratracheal instillation of iron nanoparticles in mice. Toxicology. 2010;275(1–3):65–71. doi:10.1016/j.tox.2010.06.002

197. Zhang X, Luan J, Chen W, et al. Mesoporous silica nanoparticles induced hepatotoxicity via NLRP3 inflammasome activation and caspase-1-dependent pyroptosis. Nanoscale. 2018;10(19):9141–9152. doi:10.1039/C8NR00554K

198. Hamilton RF, Wu N, Xiang C, et al. Synthesis, characterization, and bioactivity of carboxylic acid-functionalized titanium dioxide nanobelts. Part Fibre Toxicol. 2014;11:43. doi:10.1186/s12989-014-0043-7

199. Vandebriel RJ, Tonk EC, de la Fonteyne-Blankestijn LJ, et al. Immunotoxicity of silver nanoparticles in an intravenous 28-day repeated-dose toxicity study in rats. Part Fibre Toxicol. 2014;11:21. doi:10.1186/1743-8977-11-21

200. Li YJ, Wu JY, Liu J, et al. Artificial exosomes for translational nanomedicine. J Nanobiotechnology. 2021;19(1):242. doi:10.1186/s12951-021-00986-2

201. Zhang Y, Sun C, Wang C, Jankovic KE, Dong Y. Lipids and lipid derivatives for RNA delivery. Chem Rev. 2021;121(20):12181–12277. doi:10.1021/acs.chemrev.1c00244

202. Pareek A, Kumar S, Kapoor DU, Prajapati BG. Advancements in superparamagnetic nanogels: a dual-role platform for diagnosis and targeted drug delivery. Int J Pharm. 2025;677:125683. doi:10.1016/j.ijpharm.2025.125683

Continue Reading