Carnegie Mellon University Associate Professor Sufei Shi has been selected by the Gordon and Betty Moore Foundation to join its 2025 cohort of Experimental Physics Investigators. The programme will award him $1.3 million over five…
Blog
-
John Cena Leaves AJ Styles Speechless with Classy Gesture Before WWE Bout
John Cena left AJ Styles stunned with an incredible gesture honouring his career ahead of the two legend’s final match.
Cena, nearing the end of his retirement tour, personally requested to face ‘The Phenomenal One’ in one of his last encounters -…
Continue Reading
-
Chasing Michelin Stars’ Is ‘The Bear’ Made Real
Despite the way it has flooded the zone for the past few decades, I find it hard to enjoy most food television as it exists in 2025. Glitzy fine-dining porn like Chef’s Table is just that — an overwhelming series of zero-context money shots….
Continue Reading
-
Just a moment…
Just a moment… This request seems a bit unusual, so we need to confirm that you’re human. Please press and hold the button until it turns completely green. Thank you for your cooperation!
Continue Reading
-
Rinderknech upsets Medvedev to set final against cousin Vacherot, turning Shanghai into Hollywood – ATP Tour
- Rinderknech upsets Medvedev to set final against cousin Vacherot, turning Shanghai into Hollywood ATP Tour
- World No 204 Valentin Vacherot topples Djokovic to make history in Shanghai The Guardian
- Shanghai | Rinderknech springs another surprise to…
Continue Reading
-
Taylor Swift’s ‘The Life of a Showgirl’ takes aim at her friendship with Blake Lively?
Blake Lively ‘never imagined Taylor would stay upset for this long’ Taylor Swift’s latest album, The Life of a Showgirl, has seemingly shaken Blake Live to the core.
An insider told RadarOnline.com that Lively…
Continue Reading
-
PEGylated Uricase Lowers Urate of Patients With Hyperuricemia
In a phase 1, randomized, placebo-controlled, dose-escalation trial, PEGylated uricase (PEG-UOX) safely and effectively lowered serum uric acid levels over 21 days in adults with and without hyperuricemia. Results of the study, conducted by…
Continue Reading
-
Saudi delegation expresses interest to invest in Punjab – RADIO PAKISTAN
- Saudi delegation expresses interest to invest in Punjab RADIO PAKISTAN
- Aurangzeb sees new phase in Pak-Saudi economic ties Dawn
- Pakistan pitches over $28 billion ‘opportunities pipeline’ to visiting Saudi investors Arab News PK
- Saudi prince…
Continue Reading
-
Second annual swim to raise awareness of neuroendocrine cancer
Caroline RobinsonSouth West and
Hannah Staceyin Falmouth
Ruth Hitchcock
The second National Daily Dip 4 Dave Day took place on Saturday A woman in Cornwall who has completed 733 days of a 1,000 day swim challenge invited members of the public to join…
Continue Reading
-
Preparation, Characterization, and Therapeutic Applications of Plant-D
Introduction
What Do We Understand About Plant-Derived Exosome-Like Nanovesicles?
In recent years, Plant-derived exosome-like nanoparticles (PELNs) have attracted increasing attention as natural nanocarriers for biomedical applications. While mammalian-derived exosomes also demonstrate therapeutic potential, their clinical translation faces several challenges, including the risk of immune rejection, possible transmission of animal-borne pathogens, ethical concerns regarding the use of animal-derived materials, animal welfare considerations, low production yields, and the high cost of establishing large-scale culture systems. In contrast, PELNs are abundant and are typically derived from fruits, vegetables, and medicinal herbs, making them sustainable and readily available.1 They carry unique bioactive cargos such as plant-specific proteins, lipids, nucleic acids, and microRNAs(miRNAs), offering them with distinct functional properties and broad translational promise.2 Moreover, plant materials are easier to obtain, more economical to process, and simpler to store and transport, while avoiding the ethical concerns often associated with animal-derived exosomes in drug development.3
Currently, PELNs are primarily isolated using ultracentrifugation, ultrafiltration centrifugation, and density gradient centrifugation.4 However, variations in these protocols often result in substantial differences in yield, purity, and biological activity. Compared with size exclusion chromatography, ultracentrifugation allows the processing of larger sample volumes and achieves higher yields. In contrast to the more economical polymer precipitation method, it results in fewer co-precipitated impurities. Although density gradient centrifugation generally provides higher purity, both ultrafiltration centrifugation and ultracentrifugation are more practical for large-scale preparations, with ultracentrifugation often preferred for its balance of yield and feasibility (Table 1). 2,4–6 Recent efforts have therefore focused on developing scalable isolation strategies that optimize yield and purity while preserving the functional integrity of PELNs.
Table 1 Comparison of Conventional Separation Techniques for PELNs
In the characterization of PELNs, biochemical profiling serves as a critical step in distinguishing them from other types of extracellular vesicles, particularly functional microvesicles.2 Molecular characterization is commonly performed using Western blotting and flow cytometry. Potential markers for PELNs include surface proteins such as CD63,7 PEN1,8 TET8,5,8 Exo70,5 TET3,5 Class I chitinase (PR-3) and Class I β-1,3-glucanase (PR-2).9 Internal proteins such as heat shock protein 70 (HSP70), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and S-adenosylhomocysteine hydrolase (SAHH) are also frequently reported.5,8 Nucleic acids, particularly small RNAs, can be profiled using next-generation sequencing or qPCR. Lipidomic analysis via mass spectrometry has revealed that PELNs possess lipid bilayers primarily composed of phosphatidylcholine and phosphatidylethanolamine. Notably, the lipid composition varies among PELNs derived from different plant sources.7 Phosphatidic acid, in particular, is a dominant lipid species that plays a key role in the uptake and absorption of PELNs by recipient cells8 (Figure 1).
Figure 1 Structural and molecular composition of PELNs. This figure illustrates a PELN, typically 50–200 nm in diameter, with a spherical or cup-shaped morphology and a lipid bilayer membrane. Characteristic surface markers include CD63, PEN1, TET8, TET3, Exo70, Class I chitinase (PR-3), and Class I β-1,3-glucanase (PR-2). Internal contents of PELNs consist of cytosolic proteins such as HSP70, GAPDH, nucleic acids (miRNA, RNA), lipids, and other biologically active constituents. The membrane composition mainly includes phosphatidylcholine (PC), phosphatidylethanolamine (PE), and phosphatidic acid (PA), which are critical for vesicle stability and cellular uptake. The figure also includes a symbolic legend indicating the molecular components. (By Figdraw).
Due to their low immunogenicity, excellent biocompatibility, inherent targeting capacity, and suitability for surface engineering, PELNs have been widely explored in drug delivery, diagnostics, and therapeutic intervention.5,10 They have demonstrated promising effects across diverse disease models, including inflammation, oxidative stress, cancer, wound healing,8 immune regulation, neuroprotection, metabolic modulation, cardiovascular protection, gut homeostasis, osteoporosis, muscle atrophy, and premature ovarian failure. Notably, PELNs are compatible with multiple administration routes, including oral, intravenous, intratracheal, intranasal, and topical delivery.11 This review therefore provides a comprehensive overview of therapeutic applications and signaling mechanisms associated with PELNs, offering insights to guide future translational research and clinical development.
The Key Therapeutic Applications of Plant-Derived Exosome-Like Nanoparticles
Among the various biomolecules encapsulated in PELNs, miRNAs are key post-transcriptional regulators of gene expression. Their therapeutic potential lies in their ability to mediate cross-kingdom communication, thereby increasing the diversity of miRNAs in mammalian cells and exerting multi-target effects.12,13 PELNs can protect miRNAs from degradation in the gastrointestinal tract while maintaining specific concentrations. Studies suggest that PELNs contain hundreds of miRNAs, and a single miRNA can target hundreds of mRNAs. Thus, when PELNs levels reach a certain baseline, they may produce significant regulatory effects.14
For example, ginseng-derived plant exosomes carry mtr-miR159 and deliver it into bone marrow mesenchymal stem cells. This upregulates Tmem100 and activates the PI3K/Akt signaling pathway, promoting neural differentiation and enhancing peripheral nerve regeneration in a rat model of peripheral nerve injury.15 In another study, ginseng-derived exosome-like nanoparticles delivered their endogenous vvi-miR396b and ptc-miR396f into glioma cells, silencing the oncogenes c-MYC and BCL2, effectively inhibiting tumor growth and achieving efficient blood-brain barrier penetration.16
Similar to miRNA, small interfering RNA (siRNA) is a double-stranded RNA molecule approximately 20–25 nucleotides in length. It can bind perfectly to the target mRNA and induce its degradation, leading to specific gene silencing. siRNA has important potential for precision therapy, especially in cancer treatment. A typical example is ginger-derived exosome-like nanoparticles (GELNs), which deliver Bcl2 siRNA into tumor cells. By silencing the anti-apoptotic gene Bcl2, they activate the apoptosis pathway in cancer cells and significantly suppress tumor growth in a mouse breast cancer model.17
PELNs have recently been shown to influence cell fate, inflammatory responses, oxidative stress, tissue repair, metabolic regulation, and tumor immunity through diverse molecular mechanisms.18 The following studies reveal their considerable potential in disease treatment.
PELNs can regulate cell proliferation, apoptosis, differentiation, and stemness maintenance through multiple signaling pathways and key molecules, thereby contributing to tissue repair, disease therapy, and regenerative medicine. In terms of cell proliferation, PELNs from Momordica charantia are a typical example. They activate the PI3K/Akt and ERK signaling pathways, upregulate PCNA, Cyclin D1, Cyclin B1, and Ki-67, promote cell cycle progression, and enhance cell proliferation, which improves the repair capacity of cardiomyocytes after radiation injury.19 Regarding apoptosis, PELNs from Brucea javanica carry natural miRNAs (such as the let-7 family) that inhibit the PI3K/Akt/mTOR pathway while activating ROS/caspase-dependent apoptosis, leading to caspase-3 and PARP cleavage. This suppresses tumor cell survival and induces programmed cell death, highlighting the potential of plant exosomes in antitumor therapy.20 In differentiation, PELNs from Panax ginseng activate the PI3K/Akt pathway through miRNAs, significantly upregulate Nestin, β3-tubulin, NGF, BDNF, and bFGF, and drive bone marrow mesenchymal stem cells to differentiate into neurons, providing new insights into neural repair and the treatment of neurodegenerative diseases.15 In stemness maintenance, PELNs from Grape inhibit GSK-3β activity, stabilize β-catenin nuclear translocation, and activate the Wnt/β-catenin pathway. This induces transcription factors such as c-Myc, Lgr5⁺, SOX2, Nanog, OCT4, and KLF4, which enhance the self-renewal and regeneration of intestinal stem cells.21 These studies indicate that PELNs regulate cell cycle factors, apoptosis pathways, differentiation markers, and stemness-related transcription factors, thus playing important roles in cell fate and opening new directions for tissue repair, antitumor therapy, and regenerative medicine.
PELNs also exhibit strong anti-inflammatory and immunomodulatory activities. PELNs from Panax notoginseng inhibit M1 macrophage polarization and promote M2 polarization, reducing TNF-α and IL-6 while increasing IL-10, thereby alleviating inflammation.22 PELNs from Garlic suppress the TLR4/NF-κB pathway, downregulate inflammatory cytokines such as IL-6 and TNF-α, and enhance the tight junction protein ZO-1 to maintain intestinal barrier integrity.23 PELNs from Broccoli mainly act through the AMPK pathway, promoting tolerogenic dendritic cells and Tregs to restore immune homeostasis.24 These findings suggest that PELNs regulate immune cell phenotypes and cytokine levels through multiple pathways and hold therapeutic potential for inflammatory diseases.
In addition, PELNs demonstrate antioxidative potential. For example, PELNs from Mung bean sprouts activate the PI3K/Akt-Nrf2 pathway to upregulate HO-1 and SOD, and reduce oxidative stress.25 PELNs from Carrot enhance HO-1 and NQO1 via the Nrf2/ARE pathway and decrease ROS production, improving cellular antioxidant defense.26 PELNs from Ginger, which contain 6-Shogaol, also activate Nrf2 and further enhance the ability to scavenge free radicals.27 These mechanisms show that PELNs can effectively strengthen antioxidant capacity, providing new approaches for preventing tissue injury and delaying aging.
In metabolic regulation, PELNs display broad effects. PELNs from Garlic upregulate GLP-1 and IRS1/2, enhance insulin signaling, and improve glucose utilization.28 PELNs from Mung bean sprouts increase GLUT4 expression and decrease GSK-3β activity, promoting glucose uptake and glycogen synthesis and improving insulin resistance.25 PELNs from Citrus limon suppress lipid metabolism genes such as ACACA, DDHD1, and DHCR24, reduce lipid synthesis, and induce tumor cell apoptosis.29 These results indicate that PELNs can improve glucose metabolism, enhance insulin sensitivity, and regulate lipid metabolism, showing promise in the treatment of metabolic diseases.
In immune regulation and antitumor responses, PELNs demonstrate unique mechanisms. Exosomes from Artemisia annua contain plant mitochondrial DNA, which activates the cGAS-STING pathway, enhances IFN-I production, and promotes CD8⁺ T cell activation, thereby improving antitumor immunity.30 PELNs from Catharanthus roseus act through the TNF-α/NF-κB/PU.1 axis to strengthen immune cell function and relieve chemotherapy-induced immunosuppression.31 PELNs from Panax ginseng activate TLR4 signaling, drive tumor-associated macrophages toward the M1 phenotype, and enhance local immune activity.32 These studies indicate that PELNs can regulate both innate and adaptive immunity, enhance antitumor responses, and provide new strategies for cancer immunotherapy.
PELNs are currently under clinical investigation for a variety of human diseases. Ongoing clinical trials are evaluating their therapeutic efficacy in the treatment of colorectal cancer (NCT01294072), head and neck cancer (NCT01668849), and IBD treatment (NCT04879810). Table 2 and Table 3 summarize the classification of PELN-based therapies according to disease types and the therapeutic drugs delivered by PELNs, respectively.33
Table 2 Classification of PELNs Therapies by Disease Types
Table 3 Therapeutic Drugs Delivered by PELNs
Effects of Plant-Derived Exosome-Like Nanoparticles on Disease-Associated Signaling Pathways
PELNs exert therapeutic effects by modulating multiple critical signaling pathways, including PI3K/Akt, NF-κB, Wnt, AMPK, MAPK, the NLRP3 inflammasome, cGAS/STING, and Nrf2/ARE. Through these regulatory axes, PELNs influence key biological processes such as metabolic homeostasis, anti-inflammation, antioxidation, wound healing, neuroprotection, and tumor suppression, thereby offering therapeutic promise in diverse pathological conditions, including diabetes, neurodegenerative diseases, cardiovascular disorders, inflammatory diseases, and cancer.
PELNs activate PI3K/Akt pathway to promote cell survival, proliferation, and metabolic regulation. In metabolic diseases such as diabetes, PELNs enhance Glucose Transporter Type 4 (GLUT4) expression via the PI3K/Akt pathway, thereby improving insulin resistance.25 In neuroprotection (eg, neurodegeneration, ischemic stroke, and ischemia-reperfusion injury), they inhibit apoptosis and maintain blood-brain barrier (BBB) integrity.22,77,78 This pathway also facilitates wound healing by promoting skin cell proliferation, migration, extracellular matrix secretion, and angiogenesis.67 In tumors, PELNs modulate cancer cell survival, proliferation, and metabolism while inhibiting invasion and metastasis.20,58,59
NF-κB pathway is primarily involved in regulating inflammation, immune responses, and cell survival. In inflammatory diseases (eg, colitis), PELNs downregulate pro-inflammatory cytokines such as TNF-α and IL-6, alleviating inflammation.23 In bone metabolism disorders (eg, osteoporosis), they inhibit osteoclast activation to reduce bone loss.83 They also enhance immune function (eg, post-chemotherapy immunomodulation) by activating lymphocytes and macrophages,31 and contribute to anti-aging effects in skin by promoting collagen expression.72
Wnt signaling pathway regulates cell proliferation, differentiation, and tissue homeostasis. PELNs promote intestinal stem cell proliferation and differentiation via Wnt/TCF4 activation, thus supporting intestinal repair.104 In inflammatory conditions like colitis, they modulate neural stem cell differentiation in the intestine to enhance regenerative capacity.21
AMPK pathway is a master regulator of cellular energy metabolism and homeostasis. In muscle atrophy, PELNs upregulate myogenesis-related factors, enhance metabolic activity, and improve mitochondrial function.85 In inflammation (eg, colitis), they attenuate inflammation by suppressing pro-inflammatory cytokines and promoting anti-inflammatory mediators.24
MAPK pathway governs cell proliferation, differentiation, stress responses, apoptosis, and inflammation. In inflammatory liver injury (eg, acetaminophen (APAP)-induced hepatotoxicity), PELNs inhibit phosphorylation of key proteins, reducing hepatocyte apoptosis and inflammation.96 In neuroprotection and cardioprotection, they improve cell survival and prevent radiation-induced apoptosis.19,77 Additionally, they promote tissue regeneration (eg, wound healing), bone remodeling (eg, osteoporosis),67,82–84 and exert antitumor effects by suppressing proliferation, inducing apoptosis, and reducing tumor cell invasiveness.29,58
NLRP3 inflammasome regulates innate immunity and the release of pro-inflammatory cytokines. In conditions such as hepatic injury, sepsis-induced acute lung injury, and ulcerative colitis, PELNs inhibit NLRP3 inflammasome assembly, reduce cytokine release, and mitigate inflammatory damage.35,42,44,50,105
cGAS/STING pathway plays a crucial role in innate immunity, antiviral defense, and antitumor immunity. In metabolic disorders such as insulin resistance and type 2 diabetes, PELNs improve metabolic function by reducing inflammation and promoting insulin receptor substrate expression.28,76 In tumor immunoregulation, they reshape macrophage phenotypes and enhance antitumor immunity, thereby suppressing tumor growth.30
Nrf2/ARE pathway alleviates oxidative stress by upregulating antioxidant defenses. In neurodegenerative diseases (eg, Parkinson’s disease), PELNs reduce oxidative damage and enhance neuronal survival.26 In cardiovascular diseases (eg, myocardial infarction) and inflammatory liver injury (eg, alcoholic fatty liver), they suppress ROS production and induce antioxidant enzymes such as HO-1 and NQO1, thereby reducing tissue injury.26,27,79,81,106
Collectively, PELNs modulate diverse signaling pathways to regulate metabolism, inflammation, oxidative stress, tissue regeneration, and tumor progression, highlighting their broad clinical potential across multiple disease spectrums. (Figure 2).
Table 4 Summary of Signaling Pathways Affected by PELNs
Figure 2 Hierarchical representation of PELNs-regulated signaling pathways and associated diseases. This three-tier circular diagram illustrates the relationship between key signaling pathways modulated by PELNs, the major disease categories they influence, and specific pathological conditions. This three-tier circular figure illustrates the relationship between key signaling pathways modulated by plant-derived exosome-like nanoparticles (PELNs), the major disease categories they influence, and specific pathological conditions. Inner ring (core): Key signaling pathways involved in PELNs-mediated therapeutic effects, including PI3K/Akt, NF-κB, MAPK, AMPK, Wnt, NLRP3 inflammasome, cGAS/STING, Nrf2/ARE, among others. Middle ring: Broad disease categories influenced by the corresponding pathways, such as metabolic disorders, inflammatory diseases, neurodegenerative conditions, cardiovascular diseases, musculoskeletal disorders, cancers, and tissue regeneration. Outer ring: Representative diseases within each category (eg, diabetes, ulcerative colitis, ischemic stroke, myocardial infarction, osteoporosis, TNBC, etc.). The figure highlights the multifunctional regulatory roles of PELNs across diverse pathological contexts. (By Figdraw).
In addition to the major signaling pathways discussed above, several less common but biologically relevant pathways modulated by PELNs have also been identified and are comprehensively summarized in Table 4. With the continued elucidation of the molecular mechanisms by which PELNs regulate intracellular signaling, their application in precision drug delivery is expected to expand significantly. Owing to their low cytotoxicity, high biocompatibility, and minimal intrinsic immunogenicity, PELNs offer a unique therapeutic modality that integrates drug delivery, signaling modulation, and dynamic response to pathological stimuli. This multifaceted functionality enables a synergistic “delivery–regulation–therapy” strategy to enhance therapeutic efficacy. Furthermore, the inherent targeting capacity of PELNs, coupled with their surface modifiability, holds great promise for the development of intelligent drug delivery systems. Such systems would possess disease-site recognition, stimulus responsiveness, and controlled release capabilities, offering a more efficient, safe, and personalized treatment approach for chronic disorders, cancer, and inflammatory diseases.
Beyond signaling pathways, several bioactive molecules have been identified as mediators of PELN function. For example, Houttuynia cordata PELNs contain flavonoids such as luteolin,35 ginger-derived PELNs carry 6-shogaol,27 broccoli-derived PELNs deliver sulforaphane,24 and Artemisia annua PELNs contain mitochondrial DNA,30 engineered ginseng-derived ELNs are loaded with miR-182-5p,50 tomato-derived PELNs carry miR164a/b-5p,81 ginseng-derived nanoparticles contain various miRNAs,15 Momordica charantia PELNs include miR-5266 and miR-5813,78 and Brucea javanica PELNs contain functional miRNAs such as let-7.20 These findings show more clearly which bioactive components of PELNs are responsible for their therapeutic effects.
Future Prospective
With the progress of research on PELNs in drug delivery and disease treatment, their potential as novel therapeutic tools is becoming increasingly evident. Current preclinical studies have demonstrated that PELNs possess low immunogenicity, favorable biocompatibility, and can be given through multiple administration routes. They have shown positive therapeutic effects in models of inflammation, cancer, cardiovascular disease, neurodegeneration, and metabolic disorders. Importantly, PELNs can not only serve as carriers for drugs and nucleic acids (eg, miRNA and siRNA), but also provide therapeutic benefits through their own bioactive components. This dual role makes them promising for complex diseases.
In the future, several challenges still need to be addressed before PELNs can be widely used in clinical practice. First, their therapeutic effects must be reproducible and stable At present, PELNs derived from different plants may show differences in composition and function, and these differences need systematic study. Second, although animal experiments have demonstrated that PELNs can suppress inflammation, reduce oxidative stress, and promote tissue repair, their safety, effective dosage, and long-term benefits in humans remain to be clarified. Furthermore, combining PELNs with existing therapies could further improve outcomes. For example, in cancer therapy, PELNs may serve as natural carriers for nucleic acids and be used in combination with chemotherapy or immune checkpoint inhibitors, which might increase efficacy and reduce side effects.
Looking ahead, PELNs show broad prospects in treatment. They may become not only the next generation of drug delivery platforms, but also independent therapeutic agents. With further progress in preparation methods, mechanistic studies, and clinical validation, PELNs may bring new breakthroughs for the treatment of currently intractable diseases.
Funding
National Natural Science Foundation of China (82405273), Natural Science Foundation of Hubei Province (2022CFD023 and 2024AFD299). Basic scientific research project of the Educational Department of Liaoning Province (JYTMS20230584). Natural Science Foundation of Liaoning Province (2023-MSLH-028).
Disclosure
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
References
1. Zhao B, Lin H, Jiang X. et al. Exosome-like nanoparticles derived from fruits, vegetables, and herbs: innovative strategies of therapeutic and drug delivery. Theranostics. 2024;14(12):4598–4621. doi:10.7150/thno.97096
2. Yang D, Zhang W, Zhang H, et al. Progress, opportunity, and perspective on exosome isolation – efforts for efficient exosome-based theranostics[J]. Theranostics. 2020;10(8):3684–3707. doi:10.7150/thno.41580
3. Wang W, Wang P, Liang Z, et al. Exosome-like nanovesicles derived from Momordica charantia ameliorate delayed t-PA thrombolysis-induced hemorrhagic transformation by inhibiting the ONOO−/HMGB1/MMP-9 pathway. Journal of Functional Foods. 2024;114:106086. doi:10.1016/j.jff.2024.106086
4. Wang X, Xia J, Yang L, et al. Recent progress in exosome research: isolation, characterization and clinical applications[J]. Cancer Genet Ther. 2023;30(8):1051–1065. doi:10.1038/s41417-023-00617-y
5. Li Y, Wang Y, Zhao H, et al. Engineering Strategies of Plant-Derived Exosome-Like Nanovesicles: current Knowledge and Future Perspectives[J]. Int J Nanomed. 2024;19:12793–12815. doi:10.2147/IJN.S496664
6. Visan KS, Lobb RJ, Ham S, et al. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles[J]. J Extracell Vesicles. 2022;11(9):12266. doi:10.1002/jev2.12266
7. Liu Y, Ren C, Zhan R, et al. Exploring the Potential of Plant-Derived Exosome-like Nanovesicle as Functional Food Components for Human Health: a Review[J]. Foods. 2024;13(5):712. doi:10.3390/foods13050712
8. Bai C, Liu J, Zhang X, et al. Research status and challenges of plant-derived exosome-like nanoparticles[J]. Biomed Pharmacother. 2024;174:116543. doi:10.1016/j.biopha.2024.116543
9. Nguyen YNG, Pham CV, Chowdhury R, et al. Development of Blueberry-Derived Extracellular Nanovesicles for Immunomodulatory Therapy[J]. Pharmaceutics. 2023;15(8):2115. doi:10.3390/pharmaceutics15082115
10. Feng W, Teng Y, Zhong Q, et al. Biomimetic Grapefruit-Derived Extracellular Vesicles for Safe and Targeted Delivery of Sodium Thiosulfate against Vascular Calcification[J]. ACS Nano. 2023;17(24):24773–24789. doi:10.1021/acsnano.3c05261
11. Barzin M, Bagheri AM, Ohadi M, et al. Application of plant-derived exosome-like nanoparticles in drug delivery[J]. Pharmaceutical Development Technol. 2023;28(5):383–402. doi:10.1080/10837450.2023.2202242
12. Yi C, Lu L, Li Z, et al. Plant-derived exosome-like nanoparticles for microRNA delivery in cancer treatment. Drug Deliv Transl Res. 2025;15(1):84–101. doi:10.1007/s13346-024-01621-x
13. Tembo KM, Wang X, Bolideei M, et al. Exploring the bioactivity of MicroRNAs Originated from Plant-derived Exosome-like Nanoparticles (PELNs): current perspectives. J Nanobiotechnology. 2025;23(1):563. doi:10.1186/s12951-025-03602-9
14. Xu T, Zhu Y, Lin Z, et al. Evidence of Cross-Kingdom Gene Regulation by Plant MicroRNAs and Possible Reasons for Inconsistencies. J Agric Food Chem. 2024;72(9):4564–4573. doi:10.1021/acs.jafc.3c09097
15. Xu XH, Yuan TJ, Dad HA, et al. Plant Exosomes As Novel Nanoplatforms for MicroRNA Transfer Stimulate Neural Differentiation of Stem Cells In Vitro and In Vivo. Nano Lett. 2021;21(19):8151–8159. doi:10.1021/acs.nanolett.1c02530
16. Kim J, Zhu Y, Chen S, et al. Anti-glioma effect of ginseng-derived exosomes-like nanoparticles by active blood-brain-barrier penetration and tumor microenvironment modulation. J Nanobiotechnology. 2023;21(1):253. doi:10.1186/s12951-023-02006-x
17. Li Z, Wang H, Yin H, Bennett C, Zhang HG, Guo P. Arrowtail RNA for Ligand Display on Ginger Exosome-like Nanovesicles to Systemic Deliver siRNA for Cancer Suppression. Sci Rep. 2018;8(1):14644. doi:10.1038/s41598-018-32953-7
18. Wang D, Mei Z, Zhao T, et al. The roles of plant-derived nanovesicles in malignant tumours: a bibliometric analysis[J]. Int J Biol Macromol. 2025;305(Pt 2):141112. doi:10.1016/j.ijbiomac.2025.141112
19. Cui WW, Ye C, Wang KX, et al. Momordica. charantia-Derived Extracellular Vesicles-Like Nanovesicles Protect Cardiomyocytes Against Radiation Injury via Attenuating DNA Damage and Mitochondria Dysfunction[J]. Front Cardiovascular Med. 2022;9:864188. doi:10.3389/fcvm.2022.864188
20. Yan G, Xiao Q, Zhao J, et al. Brucea javanica derived exosome-like nanovesicles deliver miRNAs for cancer therapy[J]. J Control Release. 2024;367:425–440. doi:10.1016/j.jconrel.2024.01.060
21. Ju S, Mu J, Dokland T, et al. Grape Exosome-like Nanoparticles Induce Intestinal Stem Cells and Protect Mice From DSS-Induced Colitis[J]. Mol Ther. 2013;21(7):1345–1357. doi:10.1038/mt.2013.64
22. Li S, Zhang R, Wang A, et al. Panax notoginseng: derived exosome-like nanoparticles attenuate ischemia reperfusion injury via altering microglia polarization[J]. J Nanobiotechnol. 2023;21(1):416. doi:10.1186/s12951-023-02161-1
23. Zhu Z, Liao L, Gao M, et al. Garlic-derived exosome-like nanovesicles alleviate dextran sulphate sodium-induced mouse colitis via the TLR4/MyD88/NF-κB pathway and gut microbiota modulation. Food Funct. 2023;14(16):7520–7534. doi:10.1039/D3FO01094E
24. Deng Z, Rong Y, Teng Y, et al. Broccoli-Derived Nanoparticle Inhibits Mouse Colitis by Activating Dendritic Cell AMP-Activated Protein Kinase[J]. Mol Ther. 2017;25(7):1641–1654. doi:10.1016/j.ymthe.2017.01.025
25. He C, Wang K, Xia J, et al. Natural exosomes-like nanoparticles in mung bean sprouts possesses anti-diabetic effects via activation of PI3K/Akt/GLUT4/GSK-3β signaling pathway[J]. J Nanobiotechnol. 2023;21(1):349. doi:10.1186/s12951-023-02120-w
26. Kim DK, Rhee WJ. Antioxidative Effects of Carrot-Derived Nanovesicles in Cardiomyoblast and Neuroblastoma Cells[J]. Pharmaceutics. 2021;13(8):1203. doi:10.3390/pharmaceutics13081203
27. Zhuang X, Deng ZB, Mu J, et al. Ginger-derived nanoparticles protect against alcohol-induced liver damage[J]. J Extracell Vesicles. 2015;4(1):28713. doi:10.3402/jev.v4.28713
28. Sundaram K, Teng Y, Mu J, et al. Outer Membrane Vesicles Released from Garlic Exosome-like Nanoparticles (GaELNs) Train Gut Bacteria that Reverses Type 2 Diabetes via the Gut-Brain Axis[J]. Small. 2024;20(20):2308680. doi:10.1002/smll.202308680
29. Raimondo S, Saieva L, Cristaldi M, et al. Label-free quantitative proteomic profiling of colon cancer cells identifies acetyl-CoA carboxylase alpha as antitumor target of Citrus limon-derived nanovesicles[J]. J Proteomics. 2018;173:1–11. doi:10.1016/j.jprot.2017.11.017
30. Liu J, Xiang J, Jin C, et al. Medicinal plant-derived mtDNA via nanovesicles induces the cGAS-STING pathway to remold tumor-associated macrophages for tumor regression[J]. J Nanobiotechnol. 2023;21(1):78. doi:10.1186/s12951-023-01835-0
31. Ou X, Wang H, Tie H, et al. Novel plant-derived exosome-like nanovesicles from Catharanthus roseus: preparation, characterization, and immunostimulatory effect via TNF-α/NF-κB/PU.1 axis[J]. J Nanobiotechnol. 2023;21(1):160. doi:10.1186/s12951-023-01919-x
32. Cao M, Yan H, Han X, et al. Ginseng-derived nanoparticles alter macrophage polarization to inhibit melanoma growth[J]. J ImmunoTherapy Cancer. 2019;7(1):326. doi:10.1186/s40425-019-0817-4
33. Orefice NS, Di Raimo R, Mizzoni D, et al. Purposing plant-derived exosomes-like nanovesicles for drug delivery: patents and literature review[J]. Expert Opin Ther Patents. 2023;33(2):89–100. doi:10.1080/13543776.2023.2195093
34. Sriwastva MK, Deng Z, Wang B, et al. Exosome‐like nanoparticles from Mulberry bark prevent DSS‐induced colitis via the AhR/COPS8 pathway[J]. EMBO Rep. 2022;23(3):e53365. doi:10.15252/embr.202153365
35. Li JH, Xu J, Huang C, et al. Houttuynia cordata-Derived Exosome-Like Nanoparticles Mitigate Colitis in Mice via Inhibition of the NLRP3 Signaling Pathway and Modulation of the Gut Microbiota[J]. Int J Nanomed. 2024;19:13991–14018. doi:10.2147/IJN.S493434
36. Wang F, Yuan M, Shao C, et al. Momordica charantia-Derived Extracellular Vesicles Provide Antioxidant Protection in Ulcerative Colitis[J]. Molecules. 2023;28(17):6182. doi:10.3390/molecules28176182
37. Zhang M, Viennois E, Prasad M, et al. Edible ginger-derived nanoparticles: a novel therapeutic approach for the prevention and treatment of inflammatory bowel disease and colitis-associated cancer[J]. Biomaterials. 2016;101:321–340. doi:10.1016/j.biomaterials.2016.06.018
38. Zhu M, Wang S, Yue N, et al. “One stone three birds”: andrographis paniculata-derived exosome-like nanoparticles mitigate dextran sulfate sodium-induced colitis. Chin Med J. 2025;2025:10–97
39. Gong Q, Sun C, Jiang T, Guo Y. Zanthoxylum bungeanum-Derived Nanobiotics for Effective Against Ulcerative Colitis in Mouse Model. Int J Nanomed. 2025;20:6317–6331. doi:10.2147/IJN.S515961
40. Huang S, Zhang M, Li X, et al. Formulation, characterization, and evaluation of curcumin-loaded ginger-derived nanovesicles for anti-colitis activity[J]. J Pharm Anal. 2024;14(12):101014. doi:10.1016/j.jpha.2024.101014
41. Liu B, Li X, Yu H, et al. Therapeutic potential of garlic chive-derived vesicle-like nanoparticles in NLRP3 inflammasome-mediated inflammatory diseases[J]. Theranostics. 2021;11(19):9311–9330. doi:10.7150/thno.60265
42. Liu B, Lu Y, Chen X, et al. Protective Role of Shiitake Mushroom-Derived Exosome-Like Nanoparticles in D-Galactosamine and Lipopolysaccharide-Induced Acute Liver Injury in Mice[J]. Nutrients. 2020;12(2):477. doi:10.3390/nu12020477
43. Xu F, Mu J, Teng Y, et al. Restoring Oat Nanoparticles Mediated Brain Memory Function of Mice Fed Alcohol by Sorting Inflammatory Dectin-1 Complex Into Microglial Exosomes[J]. Small. 2022;18(6):2105385.
44. Chen X, Liu B, Li X, et al. Identification of anti-inflammatory vesicle-like nanoparticles in honey[J]. J Extracell Vesicles. 2021;10(4):e12069. doi:10.1002/jev2.12069
45. Naselli F, Volpes S, Cardinale S, et al. New Nanovesicles from Prickly Pear Fruit Juice: a Resource with Antioxidant, Anti-Inflammatory, and Nutrigenomic Properties[J]. Cells. 2024;13(21):1756. doi:10.3390/cells13211756
46. Urzì O, Cafora M, Ganji NR, et al. Lemon-derived nanovesicles achieve antioxidant and anti-inflammatory effects activating the AhR/Nrf2 signaling pathway[J]. iScience. 2023;26(7):107041. doi:10.1016/j.isci.2023.107041
47. Perut F, Roncuzzi L, Avnet S, et al. Strawberry-Derived Exosome-Like Nanoparticles Prevent Oxidative Stress in Human Mesenchymal Stromal Cells[J]. Biomolecules. 2021;11(1):87. doi:10.3390/biom11010087
48. Kilasoniya A, Garaeva L, Shtam T, et al. Potential of Plant Exosome Vesicles from Grapefruit (Citrus × paradisi) and Tomato (Solanum lycopersicum) Juices as Functional Ingredients and Targeted Drug Delivery Vehicles[J]. Antioxidants. 2023;12(4):943. doi:10.3390/antiox12040943
49. Kim JS, Eom JY, Kim HW, et al. Hemp sprout-derived exosome-like nanovesicles as hepatoprotective agents attenuate liver fibrosis[J]. Biomater Sci. 2024;12(20):5361–5371. doi:10.1039/D4BM00812J
50. Ma C, Liu K, Wang F, et al. Neutrophil membrane-engineered Panax ginseng root-derived exosomes loaded miRNA 182-5p targets NOX4/Drp-1/NLRP3 signal pathway to alleviate acute lung injury in sepsis: experimental studies[J]. Int J Surg. 2024;110(1):72. doi:10.1097/JS9.0000000000000789
51. Raimondo S, Naselli F, Fontana S, et al. Citrus limon -derived nanovesicles inhibit cancer cell proliferation and suppress CML xenograft growth by inducing TRAIL-mediated cell death[J]. Oncotarget. 2015;6(23):19514–19527. doi:10.18632/oncotarget.4004
52. Yang M, Liu X, Luo Q, et al. An efficient method to isolate lemon derived extracellular vesicles for gastric cancer therapy[J]. J Nanobiotechnol. 2020;18(1):100. doi:10.1186/s12951-020-00656-9
53. Zu M, Xie D, Canup BSB, et al. ‘Green’ nanotherapeutics from tea leaves for orally targeted prevention and alleviation of colon diseases[J]. Biomaterials. 2021;279:121178. doi:10.1016/j.biomaterials.2021.121178
54. Chen Q, Li Q, Liang Y, et al. Natural exosome-like nanovesicles from edible tea flowers suppress metastatic breast cancer via ROS generation and microbiota modulation[J]. Acta Pharmaceutica Sinica B. 2022;12(2):907–923. doi:10.1016/j.apsb.2021.08.016
55. Chen Q, Zu M, Gong H, et al. Tea leaf-derived exosome-like nanotherapeutics retard breast tumor growth by pro-apoptosis and microbiota modulation[J]. J Nanobiotechnol. 2023;21(1):6. doi:10.1186/s12951-022-01755-5
56. Sasaki D, Kusamori K, Takayama Y, et al. Development of nanoparticles derived from corn as mass producible bionanoparticles with anticancer activity[J]. Sci Rep. 2021;11(1):22818. doi:10.1038/s41598-021-02241-y
57. Ma X, Chen N, Zeng P, et al. Hypericum Perforatum-Derived Exosomes-Like Nanovesicles: a Novel Natural Photosensitizer for Effective Tumor Photodynamic Therapy[J]. Int J Nanomed. 2025;20:1529–1541. doi:10.2147/IJN.S510339
58. Stanly C, Alfieri M, Ambrosone A, et al. Grapefruit-Derived Micro and Nanovesicles Show Distinct Metabolome Profiles and Anticancer Activities in the A375 Human Melanoma Cell Line[J]. Cells. 2020;9(12):2722. doi:10.3390/cells9122722
59. Wang B, Guo XJ, Cai H, et al. Momordica charantia-derived extracellular vesicles-like nanovesicles inhibited glioma proliferation, migration, and invasion by regulating the PI3K/AKT signaling pathway[J]. Journal of Functional Foods. 2022;90:104968. doi:10.1016/j.jff.2022.104968
60. Luo X, Zhang X, Xu A, et al. Mechanistic Insights into the Anti-Glioma Effects of Exosome-Like Nanoparticles Derived from Garcinia Mangostana L.: a Metabolomics, Network Pharmacology, and Experimental Study. Int J Nanomed. 2025;20:5407–5427. doi:10.2147/IJN.S514930
61. Chen T, Ma B, Lu S, et al. Cucumber-Derived Nanovesicles Containing Cucurbitacin B for Non-Small Cell Lung Cancer Therapy[J]. Int J Nanomed. 2022;17:3583–3599. doi:10.2147/IJN.S362244
62. Chi Y, Shi L, Lu S, et al. Inhibitory effect of Lonicera japonica-derived exosomal miR2911 on human papilloma virus[J]. J Ethnopharmacol. 2024;318:116969. doi:10.1016/j.jep.2023.116969
63. Cui Z, Liu T, Wen Y, et al. Oral administration of cranberry-derived exosomes attenuates murine premature ovarian failure in association with changes in the specific gut microbiota and diminution in ovarian granulosa cell PANoptosis[J]. Food Funct. 2024;15(23):11697–11714. doi:10.1039/D4FO03468F
64. Qiu FS, Wang JF, Guo MY, et al. Rgl-exomiR-7972, a novel plant exosomal microRNA derived from fresh Rehmanniae Radix, ameliorated lipopolysaccharide-induced acute lung injury and gut dysbiosis[J]. Biomed Pharmacother. 2023;165:115007. doi:10.1016/j.biopha.2023.115007
65. Zou J, Song Q, Shaw PC, et al. Tangerine Peel-Derived Exosome-Like Nanovesicles Alleviate Hepatic Steatosis Induced by Type 2 Diabetes: evidenced by Regulating Lipid Metabolism and Intestinal Microflora[J]. Int J Nanomed. 2024;19:10023–10043. doi:10.2147/IJN.S478589
66. Berger E, Colosetti P, Jalabert A, et al. Use of Nanovesicles from Orange Juice to Reverse Diet-Induced Gut Modifications in Diet-Induced Obese Mice[J]. Mol Ther Methods Clin Dev. 2020;18:880–892. doi:10.1016/j.omtm.2020.08.009
67. Yang S, Lu S, Ren L, et al. Ginseng-derived nanoparticles induce skin cell proliferation and promote wound healing[J]. J Ginseng Res. 2023;47(1):133–143. doi:10.1016/j.jgr.2022.07.005
68. Tan S, Liu Z, Cong M, et al. Dandelion-derived vesicles-laden hydrogel dressings capable of neutralizing Staphylococcus aureus exotoxins for the care of invasive wounds[J]. J Control Release. 2024;368:355–371. doi:10.1016/j.jconrel.2024.02.045
69. Savcı Y, Kırbaş OK, Bozkurt BT, et al. Grapefruit-derived extracellular vesicles as a promising cell-free therapeutic tool for wound healing[J]. Food Funct. 2021;12(11):5144–5156. doi:10.1039/D0FO02953J
70. Şahin F, Koçak P, Güneş MY, et al. In Vitro Wound Healing Activity of Wheat-Derived Nanovesicles[J]. Appl. Biochem. Biotechnol. 2019;188(2):381–394. doi:10.1007/s12010-018-2913-1
71. Abraham AM, Wiemann S, Ambreen G, et al. Cucumber-Derived Exosome-like Vesicles and PlantCrystals for Improved Dermal Drug Delivery[J]. Pharmaceutics. 2022;14(3):476. doi:10.3390/pharmaceutics14030476
72. Trentini M, Zanolla I, Zanotti F, et al. Apple Derived Exosomes Improve Collagen Type I Production and Decrease MMPs during Aging of the Skin through Downregulation of the NF-κB Pathway as Mode of Action[J]. Cells. 2022;11(24):3950. doi:10.3390/cells11243950
73. Cho EG, Choi SY, Kim H, et al. Panax ginseng-Derived Extracellular Vesicles Facilitate Anti-Senescence Effects in Human Skin Cells: an Eco-Friendly and Sustainable Way to Use Ginseng Substances[J]. Cells. 2021;10(3):486. doi:10.3390/cells10030486
74. Sun Z, Zheng Y, Wang T, et al. Aloe Vera Gel and Rind-Derived Nanoparticles Mitigate Skin Photoaging via Activation of Nrf2/ARE Pathway. Int J Nanomed. 2025;20:4051–4067. doi:10.2147/IJN.S510352
75. Lee R, Ko HJ, Kim K, et al. Anti-melanogenic effects of extracellular vesicles derived from plant leaves and stems in mouse melanoma cells and human healthy skin[J]. J Extracell Vesicles. 2020;9(1):1703480. doi:10.1080/20013078.2019.1703480
76. Sundaram K, Mu J, Kumar A, et al. Garlic exosome-like nanoparticles reverse high-fat diet induced obesity via the gut/brain axis[J]. Theranostics. 2022;12(3):1220–1246. doi:10.7150/thno.65427
77. Zhang Y, Zhang X, Kai T, et al. Lycium ruthenicum Murray derived exosome-like nanovesicles inhibit Aβ-induced apoptosis in PC12 cells via MAPK and PI3K/AKT signaling pathways[J]. Int J Biol Macromol. 2024;277:134309. doi:10.1016/j.ijbiomac.2024.134309
78. Cai H, Huang LY, Hong R, et al. Momordica charantia Exosome-Like Nanoparticles Exert Neuroprotective Effects Against Ischemic Brain Injury via Inhibiting Matrix Metalloproteinase 9 and Activating the AKT/GSK3β Signaling Pathway[J]. Front Pharmacol. 2022;13:908830. doi:10.3389/fphar.2022.908830
79. Ye C, Yan C, Bian SJ, et al. Momordica charantia L.-derived exosome-like nanovesicles stabilize p62 expression to ameliorate doxorubicin cardiotoxicity[J]. J Nanobiotechnol. 2024;22(1):464. doi:10.1186/s12951-024-02705-z
80. Zhou HH, Zhou X, Pei J, et al. A fibrin gel-loaded Gouqi-derived nanovesicle (GqDNV) repairs the heart after myocardial infarction by inhibiting p38 MAPK/NF-κB p65 pathway. J Nanobiotechnology. 2025;23(1):535.
81. Shen H, Zhang M, Liu D, et al. Solanum lycopersicum derived exosome-like nanovesicles alleviate restenosis after vascular injury through the Keap1/Nrf2 pathway[J]. Food Funct. 2025;16(2):539–553. doi:10.1039/D4FO03993A
82. Cao Y, Tan X, Shen J, et al. Morinda Officinalis-derived extracellular vesicle-like particles: anti-osteoporosis effect by regulating MAPK signaling pathway[J]. Phytomedicine. 2024;129:155628. doi:10.1016/j.phymed.2024.155628
83. Seo K, Yoo JH, Kim J, et al. Ginseng-derived exosome-like nanovesicles extracted by sucrose gradient ultracentrifugation to inhibit osteoclast differentiation[J]. Nanoscale. 2023;15(12):5798–5808. doi:10.1039/D2NR07018A
84. Hwang JH, Park YS, Kim HS, et al. Yam-derived exosome-like nanovesicles stimulate osteoblast formation and prevent osteoporosis in mice[J]. J Control Release. 2023;355:184–198. doi:10.1016/j.jconrel.2023.01.071
85. Zhou X, Xu S, Zhang Z, et al. Gouqi-derived nanovesicles (GqDNVs) inhibited dexamethasone-induced muscle atrophy associating with AMPK/SIRT1/PGC1α signaling pathway[J]. J Nanobiotechnol. 2024;22(1):276. doi:10.1186/s12951-024-02563-9
86. Del Pozo-Acebo L, López de Las Hazas MC, Tomé-Carneiro J, et al. Therapeutic potential of broccoli-derived extracellular vesicles as nanocarriers of exogenous miRNAs[J]. Pharmacol Res. 2022;185:106472. doi:10.1016/j.phrs.2022.106472
87. Wang B, Zhuang X, Deng ZB, et al. Targeted Drug Delivery to Intestinal Macrophages by Bioactive Nanovesicles Released from Grapefruit[J]. Mol Ther. 2014;22(3):522–534. doi:10.1038/mt.2013.190
88. Wang Q, Zhuang X, Mu J, et al. Delivery of therapeutic agents by nanoparticles made of grapefruit-derived lipids[J]. Nat Commun. 2013;4(1):1867. doi:10.1038/ncomms2886
89. Zhang M, Xiao B, Wang H, et al. Edible Ginger-derived Nano-lipids Loaded with Doxorubicin as a Novel Drug-delivery Approach for Colon Cancer Therapy[J]. Mol Ther. 2016;24(10):1783–1796. doi:10.1038/mt.2016.159
90. You JY, Kang SJ, Rhee WJ. Isolation of cabbage exosome-like nanovesicles and investigation of their biological activities in human cells[J]. Bioact Mater. 2021;6(12):4321–4332. doi:10.1016/j.bioactmat.2021.04.023
91. Xiao Q, Zhao W, Wu C, et al. Lemon-Derived Extracellular Vesicles Nanodrugs Enable to Efficiently Overcome Cancer Multidrug Resistance by Endocytosis-Triggered Energy Dissipation and Energy Production Reduction[J]. Adv Sci. 2022;9(20):2105274. doi:10.1002/advs.202105274
92. Pomatto MAC, Gai C, Negro F, et al. Plant-Derived Extracellular Vesicles as a Delivery Platform for RNA-Based Vaccine: feasibility Study of an Oral and Intranasal SARS-CoV-2 Vaccine[J]. Pharmaceutics. 2023;15(3):974. doi:10.3390/pharmaceutics15030974
93. Mammadova R, Maggio S, Fiume I, et al. Protein Biocargo and Anti-Inflammatory Effect of Tomato Fruit-Derived Nanovesicles Separated by Density Gradient Ultracentrifugation and Loaded with Curcumin[J]. Pharmaceutics. 2023;15(2):333. doi:10.3390/pharmaceutics15020333
94. Wang Q, Ren Y, Mu J, et al. Grapefruit-Derived Nanovectors Use an Activated Leukocyte Trafficking Pathway to Deliver Therapeutic Agents to Inflammatory Tumor Sites[J]. Cancer Res. 2015;75(12):2520–2529. doi:10.1158/0008-5472.CAN-14-3095
95. Liu H, Song J, Zhou L, et al. Construction of curcumin-fortified juices using their self-derived extracellular vesicles as natural delivery systems: grape, tomato, and Orange juices[J]. Food Funct. 2023;14(20):9364–9376. doi:10.1039/D3FO02605A
96. Kim J, Gao C, Guo P, et al. A novel approach to alleviate Acetaminophen-induced hepatotoxicity with hybrid balloon flower root-derived exosome-like nanoparticles (BDEs) with silymarin via inhibition of hepatocyte MAPK pathway and apoptosis[J]. Cell Commun Signaling. 2024;22(1):334. doi:10.1186/s12964-024-01700-z
97. Sarwareddy KK, Divyash A, Sreekanth P, et al. Harnessing tomato-derived small extracellular vesicles as drug delivery system for cancer therapy[J]. Future Science OA. 2025;11(1):2461956. doi:10.1080/20565623.2025.2461956
98. Jiang D, Li Z, Liu H, et al. Plant exosome-like nanovesicles derived from sesame leaves as carriers for luteolin delivery: molecular docking, stability and bioactivity[J]. Food Chem. 2024;438:137963. doi:10.1016/j.foodchem.2023.137963
99. Ishida T, Kawada K, Jobu K, et al. Exosome-like nanoparticles derived from Allium tuberosum prevent neuroinflammation in microglia-like cells[J]. J Pharm Pharmacol. 2023;75(10):1322–1331. doi:10.1093/jpp/rgad062
100. Mao Y, Han M, Chen C, et al. A biomimetic nanocomposite made of a ginger-derived exosome and an inorganic framework for high-performance delivery of oral antibodies[J]. Nanoscale. 2021;13(47):20157–20169. doi:10.1039/D1NR06015E
101. Fang Z, Song M, Lai K, et al. Kiwi-derived extracellular vesicles for oral delivery of sorafenib[J]. Eur J Pharm Sci. 2023;191:106604. doi:10.1016/j.ejps.2023.106604
102. Sharma S, Mahanty M, Rahaman SG, et al. Avocado-derived extracellular vesicles loaded with ginkgetin and berberine prevent inflammation and macrophage foam cell formation[J]. J Cell & Mol Med. 2024;28(7):e18177. doi:10.1111/jcmm.18177
103. Li D, Yi G, Cao G, et al. Dual-Carriers of Tartary Buckwheat-Derived Exosome-Like Nanovesicles Synergistically Regulate Glucose Metabolism in the Intestine-Liver Axis[J]. Small. 2025;21:2410124.
104. Mu J, Zhuang X, Wang Q, et al. Interspecies communication between plant and mouse gut host cells through edible plant derived exosome-like nanoparticles[J]. Mol Nutr Food Res. 2014;58(7):1561–1573. doi:10.1002/mnfr.201300729
105. Chen X, Zhou Y, Yu J. Exosome-like Nanoparticles from Ginger Rhizomes Inhibited NLRP3 Inflammasome Activation[J]. Mol Pharmaceut. 2019;16(6):2690–2699. doi:10.1021/acs.molpharmaceut.9b00246
106. Gasparro R, Gambino G, Duca G, et al. Protective effects of lemon nanovesicles: evidence of the Nrf2/HO-1 pathway contribution from in vitro hepatocytes and in vivo high-fat diet-fed rats. Biomed Pharmacother. 2024;180:117532. doi:10.1016/j.biopha.2024.117532
Continue Reading