1
|
Motta MA, Martin-Saldaña S, Beloqui A, Calderón M, Larrañaga A. Polypeptide-based multilayer capsules with anti-inflammatory properties: exploring different strategies to incorporate hydrophobic drugs. J Mater Chem B 2025; 13:5297-5314. [PMID: 40207430 DOI: 10.1039/d4tb01906g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
More than 90% of drug candidates used in the drug development pipeline and about 40% of drugs on the market are poorly soluble in water based on the definition of the biopharmaceutical classification system. The advent of drug delivery approaches has represented a striking tool to overcome the challenges associated with the use of hydrophobic drugs, such as their low bioavailability and off-target effects. Drug carrier formulations composed of biodegradable and biocompatible polymers, such as polypeptides, have been explored as platforms to host poorly water-soluble drugs to prolong drug circulation, enhance their safety, reduce their immunogenicity, and promote their controlled release. In this work, we evaluated three strategies-co-precipitation, post-encapsulation, and conjugation-to incorporate a hydrophobic model drug, i.e., curcumin (CUR), into biodegradable multilayer capsules fabricated via a layer-by-layer (LbL) approach. Poly(L-lysine) (PLys) and poly(L-glutamic acid) (PGlu) were adopted as building blocks and alternately assembled onto calcium carbonate (CaCO3) microparticles to build a polypeptide-multilayer membrane, which acted as a barrier to control the release of the drug. The application of our three formulations in in vitro inflammatory models of THP-1 derived human macrophages and murine microglia showed a reduction of the inflammation with the suppression of three pivotal pro-inflammatory cytokines (i.e., interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α). Moreover, the intracellular release of CUR detected upon uptake studies on activated microglia suggested that our systems could represent a potential therapeutic approach to reduce acute neuroinflammation and modulate microglia phenotype.
Collapse
Affiliation(s)
- Maria Angela Motta
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, 48013 Bilbao, Spain.
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
| | - Ana Beloqui
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, 48013 Bilbao, Spain.
| |
Collapse
|
2
|
Liu X, Mei L, Wang J, Liu X, Yang Y, Wu Z, Ji Y. Cutting-edge insights into the mechanistic understanding of plant-derived exosome-like nanoparticles: Implications for intestinal homeostasis. Food Res Int 2025; 208:116186. [PMID: 40263791 DOI: 10.1016/j.foodres.2025.116186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/11/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
Plant-derived exosome-like nanoparticles (PDELNs) are extracted from plants such as ginger, garlic, broccoli, and others, attracting attention for their therapeutic potential due to their availability and capacity for large-scale production. Their unique physicochemical properties position PDELNs as ideal candidates for targeted gut delivery, improving intestinal health by modulating mucosal immunity, gut microbiota, and intestinal barrier integrity, all essential for maintaining intestinal homeostasis. PDELNs regulate intestinal barrier function through their bioactive components (e.g. microRNAs, lipids, and proteins). These vesicles enhance the expression of tight junction proteins and stimulate mucin production. Additionally, they promote intestinal stem cell proliferation and increase the secretion of antimicrobial peptides. PDELNs also modulate inflammatory cytokine levels and immune cell activity, fostering a balanced immune response. Further, they support the growth of beneficial gut microbiota and their metabolites, while suppressing the proliferation of pathogenic bacteria. This review summarizes recent advancements in understanding the roles of PDELNs in regulating intestinal homeostasis, focusing on their impact on mucosal immunity, intestinal barrier function, and gut microbiota composition, along with underlying molecular mechanisms and therapeutic implications. Overall, PDELNs show promise as a novel approach for treating and preventing intestinal diseases, paving the way for effective gut health interventions.
Collapse
Affiliation(s)
- Xiyuan Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Xuelian Liu
- State Key Laboratory of Direct-Fed Microbial Engineering, Beijing, 100192, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
3
|
Yuan Y, Cao K, Gao P, Wang Y, An W, Dong Y. Extracellular vesicles and bioactive peptides for regenerative medicine in cosmetology. Ageing Res Rev 2025; 107:102712. [PMID: 40032214 DOI: 10.1016/j.arr.2025.102712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
As life quality improves and the life pressure increases, people's awareness of maintaining healthy skin and hair grows. However, the use of bioactive peptides in regenerative medical aesthetics is often constrained by the high molecular weight, which impedes skin penetration. In contrast, extracellular vesicles not only possess regenerative properties but also serve as effective carriers for bioactive peptides. Given their anti-inflammatory and bactericidal properties, capacity to promote angiogenesis, optimize collagen alignment, facilitate re-epithelialization and stimulate hair growth, extracellular vesicles become an emerging and promising solution for skin regeneration treatments. The combination of peptides and extracellular vesicles enhances therapeutic efficacy and improves the bioavailability of bioactive peptides. In this review, we summarize the functions of bioactive peptides and plant- and animal-derived extracellular vesicles in regenerative medicine with cosmetology, along with examples of their combined applications. Additionally, we provide an overview of peptides and extracellular vesicles currently available on the market and in clinical practice, discussing the challenges and solutions associated with their use.
Collapse
Affiliation(s)
- Yize Yuan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Kailu Cao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Peifen Gao
- National Vaccine & Serum Institute, China National Biotech Group, Sinopharm Group, Beijing 101111, China
| | - Yinan Wang
- National Vaccine & Serum Institute, China National Biotech Group, Sinopharm Group, Beijing 101111, China
| | - Wenlin An
- National Vaccine & Serum Institute, China National Biotech Group, Sinopharm Group, Beijing 101111, China.
| | - Yiyang Dong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
4
|
Jung D, Kim NE, Kim S, Bae JH, Jung IY, Doh KW, Lee B, Kim DK, Cho YE, Baek MC. Plant-derived nanovesicles and therapeutic application. Pharmacol Ther 2025; 269:108832. [PMID: 40023319 DOI: 10.1016/j.pharmthera.2025.108832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/27/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Plant-derived nanovesicles (PDNVs) are becoming more popular as promising therapeutic tools owing to their diversity, cost-effectiveness, and biocompatibility with very low toxicity. Therefore, this review aims to discuss the methods for isolating and characterizing PDNVs and emphasize their versatile roles in direct therapeutic applications and drug delivery systems. Their ability to effectively encapsulate and deliver large nucleic acids, proteins, and small-molecule drugs was highlighted. Moreover, advanced engineering strategies, such as surface modification and fusion with other vesicles, have been developed to enhance the therapeutic effects of PDNVs. Additionally, we describe key challenges related to this field, encouraging further research to optimize PDNVs for various clinical applications for prevention and therapeutic purposes. The distinctive properties and diverse applications of PDNVs could play a crucial role in the future of personalized medicine, fostering the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Dokyung Jung
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Na-Eun Kim
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Sua Kim
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ju-Hyun Bae
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Il-Young Jung
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Kyung-Won Doh
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Do-Kyun Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong 36729, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
5
|
Rao Q, Hua H, Zhao J. Advancements in Plant-Derived sRNAs Therapeutics: Classification, Delivery Strategies, and Therapeutic Applications. Int J Mol Sci 2025; 26:4277. [PMID: 40362513 PMCID: PMC12072773 DOI: 10.3390/ijms26094277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Plant-derived small RNAs (sRNAs) have garnered significant attention in nucleic acid therapeutics, driven by their distinctive cross-kingdom regulatory capabilities and extensive therapeutic promise. These sRNAs exhibit a wide range of pharmacological effects, including pulmonary protection, antiviral, anti-inflammatory, and antitumor activities, underscoring their substantial potential for clinical translation. A key advantage lies in their delivery, facilitated by plant-specific nanovesicular carriers-such as plant exosomes, herbal decoctosomes, and bencaosomes-which protect sRNAs from gastrointestinal degradation and enable precise, tissue-specific targeting. This review provides a comprehensive analysis of plant-derived sRNAs, detailing their classification, gene-silencing mechanisms, and nanovesicle-mediated cross-kingdom delivery strategies. It further explores their therapeutic potential and underlying molecular mechanisms in major human diseases. Additionally, we critically evaluate current technical challenges and propose future directions to advance the development of plant-derived sRNAs for precision therapeutics. This work aims to offer a robust theoretical framework and practical guidance for the clinical advancement of plant-derived sRNA-based therapies.
Collapse
Affiliation(s)
- Qianru Rao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Chengdu 610041, China
| | - Junning Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Chengdu 610041, China
- National Center for Nanoscience and Technology, Beijing 100190, China
| |
Collapse
|
6
|
Zhong M, Liao T, Zeng Z, Mei J, Wu B, Lin S, Zhao Y, Tan Y, Li N, Xiu Q, Liu C, Wu X, Nie C, Lin H, Zhang Y, Li W, Li B, Pan W, Zheng L. Natural Turmeric-Derived Nanovesicles-Laden Metal-Polyphenol Hydrogel Synergistically Restores Skin Barrier in Atopic Dermatitis via a Dual-Repair Strategy. Adv Healthc Mater 2025:e2500081. [PMID: 40302269 DOI: 10.1002/adhm.202500081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/10/2025] [Indexed: 05/02/2025]
Abstract
Skin barrier impairment is critical in the development of atopic dermatitis (AD), increasing vulnerability to external pathogens and disrupting cell metabolism, which leads to inflammatory stress and immune imbalance. In this study, a natural turmeric-derived nanovesicle (TDNV)-laden metal polyphenol hydrogel, termed Fe-HD@TDNV is proposed, to synergistically restore the compromised skin barrier in AD through a dual-repair strategy. The TDNV effectively regulates metabolic activity by upregulating the expression of skin barrier proteins, antioxidant enzymes, and antimicrobial peptides (AMPs) in keratinocytes, thereby reinforcing barrier integrity and combating pathogens. Simultaneously, the Fe-HD hydrogel, cross-linked by Fe3⁺ ions and hyaluronic acid-graft-dopamine (HD), provides superior skin compatibility and establishes a low oxidative stress environment for potentiating the therapeutic efficacy of TDNV. By improving skin barrier conditions, the Fe-HD@TDNV hydrogel exhibited desirable performance in maintaining better skin hydration, reducing epidermal thickness, and decreasing abnormal immune responses in acute skin disruption models and AD models. This work is expected to offer insights into the cross-kingdom regulation between plant-derived nanovesicles and mammals, as well as the design of disease-specific dual-functional repair strategies.
Collapse
Affiliation(s)
- Mingzhen Zhong
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Tong Liao
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zehan Zeng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiangang Mei
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bodeng Wu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shan Lin
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yitao Zhao
- Department of Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third School of Clinical Medicine, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Yong Tan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ningcen Li
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qi Xiu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chunchen Liu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiuhua Wu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengtao Nie
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huixian Lin
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ye Zhang
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenbin Li
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Li
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weilun Pan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Zheng
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
7
|
Gong Q, Hu J, Pu C, Zhao Z, Guo Y. Taxus chinensis-Derived Nanovesicles Alleviate Mouse Colitis by Inhibiting Inflammatory Cytokines and Restoring Gut Microbiota. J Inflamm Res 2025; 18:5611-5625. [PMID: 40303007 PMCID: PMC12039931 DOI: 10.2147/jir.s513823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/05/2025] [Indexed: 05/02/2025] Open
Abstract
Background Recent research has increasingly focused on plant-derived products as potential alternatives to chemotherapeutic agents, aiming to reduce side effects. Among these, plant-derived nanovesicles (NVs) have garnered significant attention for their potential in treating colitis. Methods In this study, we extracted NVs from the leaves (LNVs) and stems (SNVs) of Taxus, a well-known natural anti-cancer plant. The targeting ability of these NVs was evaluated in the mouse colon using an IVIS imaging system. Additionally, we assessed the therapeutic effects of these plant-derived NVs on ulcerative colitis in a mouse model. Results Our findings reveal that the NVs exhibit an ideal vesicle size of 150.0 nm and contain a rich array of lipids, functional proteins, and bioactive small molecules. In vitro anti-inflammatory experiments demonstrated that both LNVs and SNVs enhanced cell viability and reduced levels of pro-inflammatory cytokines. Importantly, neither LNVs nor SNVs induced significant cytotoxicity. In vivo, oral administration of LNVs and SNVs ameliorated colitis-related symptoms in mice and accelerated colitis resolution by suppressing the TLR4/MyD88/NF-κB pathway and reducing levels of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Furthermore, 16S rDNA sequencing data suggested that LNVs play a crucial role in regulating gut microbiota. Conclusion Collectively, our findings suggest that plant-derived NVs from Taxus represent a promising novel natural nanomedicine for use as an anti-inflammatory agent in the treatment of colonic diseases.
Collapse
Affiliation(s)
- Qianyuan Gong
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Junqing Hu
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, 610031, People’s Republic of China
- The Center for Obesity and Metabolic Health, Department of General Surgery, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Chunlan Pu
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Zihao Zhao
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Yuanbiao Guo
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, 610031, People’s Republic of China
| |
Collapse
|
8
|
Rawat S, Arora S, Dhondale MR, Khadilkar M, Kumar S, Agrawal AK. Stability Dynamics of Plant-Based Extracellular Vesicles Drug Delivery. J Xenobiot 2025; 15:55. [PMID: 40278160 PMCID: PMC12028407 DOI: 10.3390/jox15020055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Plant-based extracellular vesicles (PBEVs) have been recognized for their wide range of applications in drug delivery however, the extent of their medicinal applicability depends on how well they are preserved and stored. Assessing their physicochemical properties, such as size, particle concentration, shape, and the activity of their cargo, forms the foundation for determining their stability during storage. Moreover, the evaluation of PBEVs is essential to ensure both safety and efficacy, which are critical for advancing their clinical development. Maintaining the biological activity of EVs during storage is a challenging task, similar to the preservation of cells and other cell-derived products like proteins. However, despite limited studies, it is expected that storing drug-loaded EVs may present fewer challenges compared to cell-based therapies, although some limitations are inevitable. This article provides a comprehensive overview of current knowledge on PBEVs preservation and storage methods, particularly focusing on their role as drug carriers. PBEVs hold promise as potential candidates for oral drug administration due to their effective intestinal absorption and ability to withstand both basic and acidic environments. However, maintaining their preservation and stability during storage is critical. Moreover, this review centers on the isolation, characterization, and storage of PBEVs, exploring the potential advantages they offer. Furthermore, it highlights key areas that require further research to overcome existing challenges and enhance the development of effective preservation and storage methods for therapeutic EVs.
Collapse
Affiliation(s)
- Satyavati Rawat
- Department of Botany, Kurukshetra University, Kurukshetra 136119, Haryana, India;
| | - Sanchit Arora
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India; (S.A.); (M.R.D.); (M.K.)
| | - Madhukiran R. Dhondale
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India; (S.A.); (M.R.D.); (M.K.)
| | - Mansi Khadilkar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India; (S.A.); (M.R.D.); (M.K.)
| | - Sanjeev Kumar
- Department of Dravyaguna, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India;
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India; (S.A.); (M.R.D.); (M.K.)
| |
Collapse
|
9
|
Lu G, Lu S, Dai H, Zhang F, Wang X, Li W, Mei L, Tan H. Engineered Turmeric-Derived Nanovesicles for Ulcerative Colitis Therapy by Attenuating Oxidative Stress and Alleviating Inflammation. Mol Pharm 2025; 22:2159-2167. [PMID: 40134348 DOI: 10.1021/acs.molpharmaceut.4c01328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Inflammation and oxidative stress are important features of traumatic ulcerative colitis (UC). Turmeric has been used as a dietary and functional ingredient for its potent anti-inflammatory effects in UC therapy. However, its practical effectiveness is hindered by limited reactive oxygen species (ROS) elimination properties. To address this, we constructed a unique treatment agent by growing cerium oxide (CeO2) nanocrystals on the membranes of turmeric-derived nanovesicles (TNVs), named as TNV-Ce. The resulted TNV-Ce could suppress inflammation and exhibit exceptional ROS-scavenging activity, which was validated both in lipopolysaccharide-induced macrophages and dextran sulfate sodium salt-induced chronic colitis mouse model. Following oral administration, TNV-Ce significantly accumulated at inflamed sites, effectively eliminating ROS and inhibiting pro-inflammatory cytokines for synergistic action against UC.
Collapse
Affiliation(s)
- Guihong Lu
- Department of Neurosurgery, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
- Center for Child Care and Mental Health (CCCMH), Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Shanming Lu
- Department of Pathology, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Haibing Dai
- Department of Pathology, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Fan Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
- Department of Pathology, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Xiaotian Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Huzhou, Zhejiang 313201, China
| | - Weiqun Li
- Department of Pathology, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Hui Tan
- Center for Child Care and Mental Health (CCCMH), Shenzhen Children's Hospital, Shenzhen 518038, China
| |
Collapse
|
10
|
Gao C, Yang Z, Song R, Sheng H, Zhu L. Nanotechnology-based drug delivery system for targeted therapy of ulcerative colitis from traditional Chinese medicine: A review. Int J Pharm 2025; 673:125375. [PMID: 39965734 DOI: 10.1016/j.ijpharm.2025.125375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disease and seriously affects the normal life of patients. Conventional therapeutic drugs are difficult to meet clinical needs. Traditional Chinese medicine (TCM) ingredients could effectively alleviate the symptoms of UC by anti-inflammatory, anti-oxidative, regulating the gut microbiota, and repairing the colonic epithelial barrier, but their low solubility and bioavailability severely limit their clinical application. Nano-drug delivery systems (NDDS) combined with TCM ingredients is a promising option for treating UC, and they could significantly enhance the stability, solubility, and bioavailability of TCM ingredients. The review describes the anti-UC mechanisms of TCM ingredients, systematically summarizes various kinds of NDDS for TCM ingredients according to different routes of administration, and highlights the advantages of NDDS for TCM ingredients in the treatmentof UC. In addition, we discuss the limitations of existing NDDS for TCM ingredients and the development direction in the future. This review will provide a basis for the future development of anti-UC NDDS for TCM ingredients.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zerun Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruirui Song
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
11
|
Han Y, Guo X, Ji Z, Guo Y, Ma W, Du H, Guo Y, Xiao H. Colon health benefits of plant-derived exosome-like nanoparticles via modulating gut microbiota and immunity. Crit Rev Food Sci Nutr 2025:1-21. [PMID: 40105379 DOI: 10.1080/10408398.2025.2479066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Exosomes are nanoscale particles with a lipid bilayer membrane that were first identified in mammalian cells. Plant-derived exosome-like nanoparticles (PELNs) share structural and molecular similarities with mammalian exosomes, including lipids, proteins, microRNA (miRNA), and plant-derived metabolites. Owing to their unique characteristics, such as outstanding stability, low immunogenicity, high biocompatibility, and sustainability, PELNs have emerged as promising natural bioactive agents with the capacity for cross-kingdom cellular regulation. Dietary supplementation with PELNs, particularly from fruits and vegetables, has demonstrated health benefits. An increasing number of studies have indicated the beneficial effects of PELNs on colon health. This review summarizes the isolation and characterization of PELNs, and their stability, uptake, and distribution after oral ingestion. Furthermore, this review emphasizes the interactions between PELNs, gut microbiota, and the gut immune system, including the uptake of PELNs by gut microbiota, modulation of gut bacteria metabolism, and immune responses by PELNs. Additionally, the applications of PELNs as bioactive components and drug carriers targeting the colon are reviewed. In summary, PELNs represent a versatile and natural approach to improve colon health, with potential applications in both therapeutic and preventive healthcare strategies.
Collapse
Affiliation(s)
- Yanhui Han
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Xiaojing Guo
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Zhengmei Ji
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
| | - Yuxin Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, PR China
| | - Wenjun Ma
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Yurong Guo
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, PR China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an, PR China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
12
|
Su X, Wang H, Li Q, Chen Z. Extracellular Vesicles: A Review of Their Therapeutic Potentials, Sources, Biodistribution, and Administration Routes. Int J Nanomedicine 2025; 20:3175-3199. [PMID: 40098717 PMCID: PMC11913029 DOI: 10.2147/ijn.s502591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Extracellular vesicles (EVs) participate in intercellular communication and play an essential role in physiological and pathological processes. In recent years, EVs have garnered significant attention as cell-free therapeutic alternatives, vectors for drug and gene delivery, biomarkers for disease diagnosis and prognosis, vaccine development, and nutraceuticals. The biodistribution of EVs critically influences their efficacy and toxicity. Therefore, this review aims to discuss the main factors influencing the biodistribution of unmodified EVs, highlighting their distribution patterns, advantages, limitations, and applications under different routes of administration. In addition, we provide a comprehensive discussion of the currently available sources of EVs and summarize the current status of the therapeutic potentials of EVs. By optimizing administration routes and selecting appropriate EV sources, we aim to offer valuable insights to enhance the delivery efficiency and therapeutic efficacy of EVs to target tissues.
Collapse
Affiliation(s)
- Xiaorong Su
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Hongxiang Wang
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, People’s Republic of China
| | - Qiubai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Engineering Research Center for Application of Extracellular Vesicle, Hubei University of Science and Technology, Xianning, 437100, People’s Republic of China
| | - Zhichao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
13
|
Wang F, Guo SQ, Su TH, Tian XJ, Wen WJ, Pan HP, Wang XF, Zhang W, Zhong JL, Dong ZS, Luo P. Bioactive-Enriched Nanovesicles from American Cockroaches Enhance Wound Healing by Promoting Angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13565-13576. [PMID: 39988799 DOI: 10.1021/acsami.4c21532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Skin trauma often results from pain, swelling, and scarring and can significantly interfere with daily activities. Extracts from the American cockroach, a rapidly reproducing insect, have been recognized for therapeutic properties in wound management. Traditional extraction methods use solvents such as ethanol to obtain the active compounds, but these methods may compromise the intrinsic biological properties of American cockroach extracts. In this study, we investigated the use of nanovesicles isolated from fresh American cockroaches in skin wound treatment and focused on their biological characteristics and therapeutic efficacy. Fresh and dried American cockroach nanovesicles (F-ACNVs and D-ACNVs, respectively) were procured via ultrahigh-speed centrifugation. We found that F-ACNVs exhibited superior cell proliferation-promoting activity. By employing metabolomics, proteomics, and long noncoding RNA (lncRNA) omics, we identified a rich repertoire of metabolites, proteins, and lncRNAs within F-ACNVs. In vitro and in vivo experiments demonstrated that F-ACNVs significantly enhanced the proliferation and migration of human umbilical vein endothelial cells (HUVECs) and human skin keratinocytes (HACATs) as well as the repair of skin mechanical trauma. These effects may be mediated through the activation of angiogenic signaling pathways. Our research introduces a novel therapeutic strategy for treating skin trauma and offers insight into the medicinal potential of insects such as the American cockroach while emphasizing the importance of preserving the intrinsic biological properties of insects for optimal therapeutic outcomes.
Collapse
Affiliation(s)
- Fang Wang
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Shi-Qi Guo
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Tao-Hong Su
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiu-Jia Tian
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wei-Jie Wen
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Hua-Ping Pan
- Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiao-Fen Wang
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wen Zhang
- Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing-Li Zhong
- Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Zi-Shu Dong
- Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ping Luo
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| |
Collapse
|
14
|
Njoku GC, Forkan CP, Soltysik FM, Nejsum PL, Pociot F, Yarani R. Unleashing the potential of extracellular vesicles for ulcerative colitis and Crohn's disease therapy. Bioact Mater 2025; 45:41-57. [PMID: 39610953 PMCID: PMC11602541 DOI: 10.1016/j.bioactmat.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- George Chigozie Njoku
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, USA
| | - Cathal Patrick Forkan
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Pharmacy, Université Grenoble Alpes, France
| | - Fumie Mitani Soltysik
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Peter Lindberg Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| |
Collapse
|
15
|
Muhammad Z, Muhammad SA, Abbas AY, Achor M, Adeyemi SA, Choonara YE, Saidu Y, Bilbis LS. Isolation and characterization of medicinal plant-based extracellular vesicles as nano delivery systems for ascorbic acid. J Microencapsul 2025; 42:120-131. [PMID: 39716732 DOI: 10.1080/02652048.2024.2443430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
AIM Plant-derived extracellular vesicles (EVs) are natural nanovesicles for drug delivery. This study isolated and characterised EVs from medicinal plants as delivery vehicles. METHODS Precipitation method was employed for the isolation and characterised using DLS, SEM, and TEM. The encapsulation efficiency (EE) and antioxidant activity of ascorbic acid (AA)-EVs were evaluated. RESULTS The total yields of lyophilised vesicles per weight of the sample were 6.0, 8.6 and 9.2 mg/g for garlic, turmeric and ginger, respectively. Mean size of garlic-derived EVs, ginger-derived EVs, and turmeric-derived EVs were 101.0 ± 6.7, 226.4 ± 62.2 and 90.7 ± 2.5 nm, respectively. The zeta potential of the EVs was between -33.2 ± 10.9 and -28.8 ± 8.43 mV. Spherical morphology of the nanovesicles was confirmed by SEM and TEM. The EE of the EVs was between 78.1 ± 2.8% and 87.2 ± 1.4%. CONCLUSION Overall, the antioxidant activity of AA-loaded EVs was better compared to free AA. This study provides evidence that these medicinal plants are rich sources for developing nanotherapeutics.
Collapse
Affiliation(s)
- Zainab Muhammad
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Suleiman A Muhammad
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Abdullahi Y Abbas
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Mohammed Achor
- Department of Pharmaceutics and Pharmaceutical Technology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Samson A Adeyemi
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Yusuf Saidu
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Lawal S Bilbis
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| |
Collapse
|
16
|
Wang R, Zhang Y, Guo Y, Zeng W, Li J, Wu J, Li N, Zhu A, Li J, Di L, Cao P. Plant-derived nanovesicles: Promising therapeutics and drug delivery nanoplatforms for brain disorders. FUNDAMENTAL RESEARCH 2025; 5:830-850. [PMID: 40242551 PMCID: PMC11997602 DOI: 10.1016/j.fmre.2023.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/17/2023] [Accepted: 09/10/2023] [Indexed: 04/18/2025] Open
Abstract
Plant-derived nanovesicles (PDNVs), including plant extracellular vesicles (EVs) and plant exosome-like nanovesicles (ELNs), are natural nano-sized membranous vesicles containing bioactive molecules. PDNVs consist of a bilayer of lipids that can effectively encapsulate hydrophilic and lipophilic drugs, improving drug stability and solubility as well as providing increased bioavailability, reduced systemic toxicity, and enhanced target accumulation. Bioengineering strategies can also be exploited to modify the PDNVs to achieve precise targeting, controlled drug release, and massive production. Meanwhile, they are capable of crossing the blood-brain barrier (BBB) to transport the cargo to the lesion sites without harboring human pathogens, making them excellent therapeutic agents and drug delivery nanoplatform candidates for brain diseases. Herein, this article provides an initial exposition on the fundamental characteristics of PDNVs, including biogenesis, uptake process, isolation, purification, characterization methods, and source. Additionally, it sheds light on the investigation of PDNVs' utilization in brain diseases while also presenting novel perspectives on the obstacles and clinical advancements associated with PDNVs.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingjie Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yumiao Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wei Zeng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jinge Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jie Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Anran Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
17
|
Gao C, Chen Y, Wen X, Han R, Qin Y, Li S, Tang R, Zhou W, Zhao J, Sun J, Li Z, Tan Z, Wang D, Zhou C. Plant-derived exosome-like nanoparticles in tissue repair and regeneration. J Mater Chem B 2025; 13:2254-2271. [PMID: 39817682 DOI: 10.1039/d4tb02394c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
This article reviews plant-derived exosome-like nanoparticles (ELNs), and highlights their potential in regenerative medicine. Various extraction techniques, including ultracentrifugation and ultrafiltration, and their impact on ELN purity and yield were discussed. Characterization methods such as microscopy and particle analysis are found to play crucial roles in defining ELN properties. This review is focused on exploring the therapeutic potential of ELNs in tissue repair, immune regulation, and antioxidant activities. Further research and optimization methods for extraction of ELNs to realize clinical potential applications are necessary.
Collapse
Affiliation(s)
- Canyu Gao
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Yang Chen
- Center of Medical Product Technical Inspection, Chengdu, 610015, China
| | - Xingyue Wen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ruiying Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuxiang Qin
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Sijie Li
- Department of Burn and Plastic Reconstructive Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Weikai Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junyu Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhengyong Li
- Department of Burn and Plastic Reconstructive Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Deli Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Changchun Zhou
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
18
|
Liu H, Liu Y, Peng S, Zhou L, McClements DJ, Fang S, Liu W. Colonic delivery and controlled release of curcumin encapsulated within plant-based extracellular vesicles loaded into hydrogel beads. Food Res Int 2025; 202:115540. [PMID: 39967127 DOI: 10.1016/j.foodres.2024.115540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 11/14/2024] [Accepted: 12/28/2024] [Indexed: 02/20/2025]
Abstract
In this study, the application of banana-derived extracellular vesicles (EVs) as natural carriers for hydrophobic nutraceuticals was investigated for the first time. Curcumin was solubilized in an amorphous state within the hydrophobic domains of the EVs in banana juice. The bioaccessibility of the curcumin was considerably higher in the curcumin-loaded EVs (CEVs) than for pure curcumin crystals. The retention/release behavior of the curcumin in the CEVs was regulated by incorporating the curcumin-loaded banana juice into hydrogel beads, which were assembled from sodium alginate and banana pectin. The pectin and D-galacturonic acid contents of the banana juice were 0.87 % ± 0.11 % and 0.58 % ± 0.08 %, respectively. With the introduction of sodium alginate, the sphericity, hardness, chewiness, water holding capacity, surface smoothness and other physiochemical properties of the hydrogel beads were improved. Fourier Transform infrared analysis showed that hydrogen bonding played an important role in the formation of the hydrogel beads. The hydrogel beads had a higher swelling ratio under neutral conditions than under acidic conditions. Encapsulation of the curcumin-loaded banana juice within the hydrogel beads retarded curcumin release and enhanced the therapy potential of ulcerative colitis, which indicated that these delivery systems could be designed to regulate the release profile of the CEVs. Encapsulation of the curcumin also improved its resistance to heating, exposure and storage. In summary, we have shown that encapsulation of curcumin within plant-based EVs loaded into hydrogel beads can improve its functional performance.
Collapse
Affiliation(s)
- Hang Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Yikun Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Shengfeng Peng
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang 330006, PR China.
| | - Lei Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - David Julian McClements
- Biopolymers and Colloids Laboratory, Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Suqiong Fang
- Sirio Pharma Co., Ltd., Shantou, Guangdong 515041, PR China
| | - Wei Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, PR China.
| |
Collapse
|
19
|
Li Y, Shao S, Zhou Y, Wang Y, Zheng W, Wang H, Wang M, Jin K, Zou H, Mou X. Oral administration of Folium Artemisiae Argyi-derived exosome-like nanovesicles can improve ulcerative colitis by regulating intestinal microorganisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156376. [PMID: 39813847 DOI: 10.1016/j.phymed.2025.156376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Ulcerative colitis (UC), an inflammatory disease characterized by intestinal barrier dysfunction, poses significant challenges because of the toxicity and adverse effects commonly associated with conventional therapies. Safer and more efficacious treatment strategies are needed. PURPOSE The purpose of this study was to treat UC with Folium Artemisiae Argyi exosome-like nanovesicles (FAELNs) and to explore its related mechanism to provide a safer and more effective means for the treatment of ulcerative colitis. METHODS We established an in vivo model of acute UC in mice and an in vitro inflammatory model using HT-29 human colorectal cancer cells. To evaluate the therapeutic effect of FAELNs on UC, we adopted various proxies, including changes in body weight and disease activity index (DAI) of mice, and measurement of colon length. The concentrations of myeloperoxide, interleukin (IL-1β), IL-6, tumor necrosis factor-alpha, monocyte chemoattractant protein-1, and interferon-gamma in sera of mice were detected by ELISA. Immunohistochemistry, hematoxylin and eosin staining, and Alyssin blue staining were performed. The effect of HT-29 cells on oxidative stress was detected using an active oxygen probe, diacetyldichlorofluorescein, and flow cytometry. Western blotting was performed to detect the expression levels of Bax and Bcl-2 in HT-29 cells treated with FAELNs. The effects of FAELNs on IL-6 and IL-1β were detected by fluorescence quantitative PCR. Fecal 16S bacteria were detected, and the role of FAELNs was verified by α diversity and β diversity analyses, principal component analysis, species distribution, and function prediction. For microRNA sequencing of FAELNs, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed. To detect the metabolic and lipid groups of FAELNs, the components were identified and a pharmacological network was constructed to explore the related mechanisms and diseases. RESULTS FAELNs effectively alleviated the pathogenesis of UC induced by dextran sodium sulfate in animal models, restoring the integrity of the intestinal barrier and reversing an imbalance of the intestinal microbiota. CONCLUSION Our findings demonstrate the therapeutic potential of FAELNs in UC management, highlighting their scalability for mass production and encouraging prospects for clinical transformation.
Collapse
Affiliation(s)
- Yishu Li
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Su Shao
- Department of General Surgery, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou 311700, PR China
| | - Yuanhao Zhou
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China
| | - Yuanyuan Wang
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China
| | - Wenjie Zheng
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Huanying Wang
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou 310012, PR China
| | - Meixia Wang
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou 310012, PR China; EVitai Bio (Hangzhou) Co. Ltd, Hangzhou 310056, PR China
| | - Ketao Jin
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310003, PR China.
| | - Hai Zou
- Department of Critical Care, Shanghai Cancer Center, Fudan University, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China.
| | - Xiaozhou Mou
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, PR China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
20
|
Li J, Luo T, Wang D, Zhao Y, Jin Y, Yang G, Zhang X. Therapeutic application and potential mechanism of plant-derived extracellular vesicles in inflammatory bowel disease. J Adv Res 2025; 68:63-74. [PMID: 38341033 PMCID: PMC11785581 DOI: 10.1016/j.jare.2024.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/09/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Plant-derived extracellular vesicles (PDEVs) are membrane vesicles characterized by a phospholipid bilayer as the basic skeleton that is wrapped by various functional components of proteins and nucleic acids. An increasing number of studies have confirmed that PDEVs can be a potential treatment of inflammatory bowel disease (IBD) and can, to some extent, compensate for the limitations of existing therapies. AIM OF REVIEW This review summarizes the recent advances and potential mechanisms underlying PDEVs obtained from different sources to alleviate IBD. In addition, the review discusses the possible applications and challenges of PDEVs, providing a theoretical basis for exploring novel and practical therapeutic strategies for IBD. KEY SCIENTIFIC CONCEPTS OF REVIEW In IBD, the crosstalk mechanism of PDEVs may regulate the intestinal microenvironment homeostasis, especially immune responses, the intestinal barrier, and the gut microbiota. In addition, drug loading enhances the therapeutic potential of PDEVs, particularly regarding improved tissue targeting and stability. In the future, not only immunotherapy based on PDEVs may be an effective treatment for IBD, but also the intestinal barrier and intestinal microbiota will be a new direction for the treatment of IBD.
Collapse
Affiliation(s)
- Jinling Li
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, Zhejiang Province, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China
| | - Ting Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China
| | - Yao Zhao
- Biomanufacturing Research Institute of Xianghu Laboratory, Hangzhou 311231, Zhejiang Province, China
| | - Yuanxiang Jin
- Biomanufacturing Research Institute of Xianghu Laboratory, Hangzhou 311231, Zhejiang Province, China; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang Province, China
| | - Guiling Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang Province, China; Biomanufacturing Research Institute of Xianghu Laboratory, Hangzhou 311231, Zhejiang Province, China.
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, Zhejiang Province, China.
| |
Collapse
|
21
|
Tang X, He M, Ren Y, Ji M, Yan X, Zeng W, Lv Y, Li Y, He Y. Traditional Chinese Medicine formulas-based interventions on colorectal carcinoma prevention: The efficacies, mechanisms and advantages. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:119008. [PMID: 39471879 DOI: 10.1016/j.jep.2024.119008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/08/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Traditional Chinese Medicine Formulas (TCMFs) represent a distinctive medical approach to disease treatment and have been utilized in clinical practice for treating intestinal diseases for thousands of years. Recently, TCMFs have received increasing attention due to their advantages of high efficiency, safety, as well as low toxicity, providing promising strategies for preventing colorectal carcinoma (CRC). Nonetheless, the potential mechanism of TCMFs in preventing CRC has not been fully elucidated. AIM OF THE STUDY The literature from the past three years was reviewed to highlight the therapeutic effects and underlying mechanisms of TCMFs in preventing CRC. MATERIALS AND METHODS The keywords have been searched, including "traditional Chinese medicine formulas," "herb pairs," "Herbal plant-derived nanoparticles," et al. in "PubMed" and "China National Knowledge Infrastructure (CNKI)," and screened published articles related to the treatment of intestinal precancerous lesions. This review primarily examined the effectiveness and mechanisms of TCMFs in treating intestinal precancerous lesions, highlighting their significant potential in preventing CRC. RESULTS Gegen Qinlian decoction, Shaoyao decoction, Wu Wei Wan, etc., exert substantial therapeutic effects on intestinal precancerous lesions. These therapeutic effects are demonstrated by a reduction in disease activity index scores, suppression of intestinal inflammation, and preservation of body weight and intestinal function, all of which contribute to the effective prevention of CRC. Besides, the classic Chinese herbal pairs and the extracellular vesicle-like nanoparticles of herbaceous plants have demonstrated superior efficacy in the treatment of intestinal precancerous lesions. Mechanistically, protecting the epithelial barrier, regulating gut microbiota as well as related metabolism, modulating macrophage polarization, and maintaining immune balance contribute to the role of TCMFs in CRC prevention. CONCLUSIONS This review demonstrates the great potential and mechanism of TCMFs in CRC prevention and provides a scientific basis for their utilization in CRC prevention.
Collapse
Affiliation(s)
- Xiaojuan Tang
- School of biomedical sciences, Hunan University, Changsha, 410012, Hunan, China; Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China.
| | - Min He
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yuan Ren
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Meng Ji
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xiaoqi Yan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China
| | - Wen Zeng
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yuan Lv
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China
| | - Yongmin Li
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China
| | - Yongheng He
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine (The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine), Changsha, 410006, Hunan, China; Hunan Academy of Chinese Medicine, Changsha, 410006, Hunan, China; Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
22
|
Jiang HY, Shao B, Wang HD, Zhao WQ, Ren SH, Xu YN, Liu T, Sun CL, Xiao YY, Li YC, Chen Q, Zhao PY, Yang GM, Liu X, Ren YF, Wang H. Analysis of nanomedicine applications for inflammatory bowel disease: structural and temporal dynamics, research hotspots, and emerging trends. Front Pharmacol 2025; 15:1523052. [PMID: 39845796 PMCID: PMC11750799 DOI: 10.3389/fphar.2024.1523052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/24/2024] [Indexed: 01/24/2025] Open
Abstract
Background The application of nanomedicine in inflammatory bowel disease (IBD) has gained significant attention in the recent years. As the field rapidly evolves, analyzing research trends and identifying research hotpots are essential for guiding future advancements, and a comprehensive bibliometric can provide valuable insights. Methods The current research focused on publications from 2001 to 2024, and was sourced from the Web of Science Core Collection (WoSCC). CiteSpace and VOSviewer were employed to visualize authors, institutions, countries, co-cited references, and keywords, thereby mapping the intellectual structure and identifying emerging trends in the field. Results The analysis covered 1,518 literature across 447 journals, authored by 9,334 researchers from 5,459 institutions and 287 countries/regions. The global publication numbers exhibited an upward trend, particularly in the last decade, with China leading as the top publishing country and the Chinese Academy of Sciences emerging as the foremost institution. Dr. Xiao Bo is the prominent figure in advanced drug delivery systems. This interdisciplinary field, which spans materials science, pharmacy, and medicine, has seen influential publications mainly concentrated on targeted nanoparticles treatment for IBD. Keyword analysis revealed that current research hotspots include drug delivery, immune cell regulation, antioxidant damage, intestinal microbiota homeostasis, and nanovesicles. Conclusion This study offers a comprehensive overview of global research landscape, emphasizing the rapid growth and increasing complexity of this field. It identifies key research hotspots and trends, including efforts to enhance the precision, efficacy, and safety of nanomedicine applications. Emerging directions are highlighted as crucial for further progress in this evolving area.
Collapse
Affiliation(s)
- Hong-Yu Jiang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong-Da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen-Qi Zhao
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| | - Shao-Hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yi-Ni Xu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Cheng-Lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Yi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Cheng Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng-Yu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Guang-Mei Yang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu-Fan Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| |
Collapse
|
23
|
Li L, Zhong D, Wang S, Zhou M. Plant-derived materials for biomedical applications. NANOSCALE 2025; 17:722-739. [PMID: 39605132 DOI: 10.1039/d4nr03057e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
With exceptional biocompatibility and biodegradability, plant-derived materials have garnered significant interest for a myriad of biomedical applications. This mini-review presents a concise overview of prevalent plant-derived materials, encompassing polysaccharide-based polymers, protein-based polymers, extracellular vesicles, mucilage, decellularized scaffolds, and whole plant-based biomass. Through different processing techniques, these plant-derived materials can be tailored into a variety of forms, such as nanoparticles, nanofibers, and hydrogels, to address the nuanced requirements of biomedical applications. With the emphasis on wound healing, tissue engineering, and drug delivery, this review underscores the unique advantages of plant-derived materials, such as lower risk of endotoxin and virus contamination, reduced ethical concerns, scalability, and eco-friendly attributes. However, challenges such as the need for the development of standardized isolation methods of these materials, and further transition from preclinical to clinical applications still remain to be solved.
Collapse
Affiliation(s)
- Lele Li
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China.
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
- Zhejiang University-Ordos City Etuoke Banner Joint Research Center, 314400, Haining, China
| | - Danni Zhong
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China.
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
- Zhejiang University-Ordos City Etuoke Banner Joint Research Center, 314400, Haining, China
| | - Shoujie Wang
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China.
| | - Min Zhou
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000, Yiwu, China.
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
- Zhejiang University-Ordos City Etuoke Banner Joint Research Center, 314400, Haining, China
- The National Key Laboratory of Biobased Transportation Fuel Technology, Zhejiang University, 310027, Hangzhou, China
| |
Collapse
|
24
|
Guo F, Wu Y, Wang G, Liu J. Role of PCBP2 in regulating nanovesicles loaded with curcumin to mitigate neuroferroptosis in neural damage caused by heat stroke. J Nanobiotechnology 2024; 22:800. [PMID: 39731111 DOI: 10.1186/s12951-024-02889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/01/2024] [Indexed: 12/29/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the mechanisms by which nanovesicles (NVs) transport curcumin(CUR) across the blood-brain barrier to treat hypothalamic neural damage induced by heat stroke by regulating the expression of poly(c)-binding protein 2 (PCBP2). METHODS Initially, NVs were prepared from macrophages using a continuous extrusion method. Subsequently, CUR was loaded into NVs using sonication, yielding engineered cell membrane Nanovesicles loaded with curcumin (NVs-CUR), which were characterized and subjected to in vitro and in vivo tracking analysis. Evaluations included assessing the toxicity of NVs-CUR using the MTT assay, evaluating neuroprotection of NVs-CUR against H2O2-induced oxidative stress damage in PC12 cells, examining effects on cell morphology and quantity, and detecting ferroptosis-related markers through Western blot and transmission electron microscopy (TEM). Proteomic analysis was conducted on PC12 cells treated with NVs (n = 3) and NVs-CUR (n = 3) to identify downstream key factors. Subsequently, the expression of key factors was modulated, and rescue experiments were performed to validate the impact of NVs-CUR through the regulation of key factor expression. Furthermore, a mouse model of hypothalamic neural damage induced by heat stroke was established, where CUR, NVs-CUR, and ferroptosis inducer Erastin were administered to observe mouse survival rates, conduct nerve function deficit scoring, perform histological staining, and measure levels of inflammation and oxidative stress factors in hypothalamic tissue. RESULTS NVs-CUR was successfully prepared with excellent stability, serving as an advantageous drug delivery system that effectively targets brain injury sites or neurons both in vitro and in vivo. Subsequent in vitro cell experiments demonstrated the biocompatibility of NVs-CUR, showing superior protective effects against H2O2-induced PC12 cell damage and reduced ferroptosis compared to CUR. Moreover, in the mouse model of hypothalamic neural damage induced by heat stroke, NVs-CUR exhibited enhanced therapeutic effects. Proteomic analysis revealed that NVs-CUR exerted its effects through the regulation of key protein PCBP2; silencing PCBP2 reversed the protective effect of NVs-CUR on neural damage and its inhibition of ferroptosis. Additionally, NVs-CUR regulated solute carrier family 7 member 11 (SLC7A11) expression by PCBP2; overexpression of SLC7A11 reversed the promotion of neural damage and ferroptosis by silencing PCBP2. Animal experiments indicated that ferroptosis inducers reversed the improved survival and nerve function observed with NVs-CUR, silencing PCBP2 reversed the ameliorative effects of NVs-CUR on hypothalamic neural injury induced by heat stroke, and overexpression of SLC7A11 further reversed the adverse effects of silencing PCBP2 on hypothalamic neural injury induced by heat stroke. This suggests that NVs-CUR alleviates hypothalamic neural damage induced by heat stroke by targeting the PCBP2/SLC7A11 axis to reduce neuronal ferroptosis. CONCLUSION This study successfully developed engineered cell membrane NVs-CUR with neuron-targeting properties. NVs-CUR increased the expression of PCBP2, maintained the stability of SLC7A11 mRNA, reduced ferroptosis, and ultimately alleviated hypothalamic neuroinflammation induced by heatstroke.
Collapse
Affiliation(s)
- Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yizhan Wu
- Graduate School of Xinjiang Medical University, Urumqi, China
| | - Guangjun Wang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, No. 359, Youhao North Road, Urumqi, Xinjiang, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, No. 359, Youhao North Road, Urumqi, Xinjiang, China.
| |
Collapse
|
25
|
Li JH, Xu J, Huang C, Hu JX, Xu HM, Guo X, Zhang Y, Xu JK, Peng Y, Zhang Y, Zhu MZ, Zhou YL, Nie YQ. Houttuynia cordata-Derived Exosome-Like Nanoparticles Mitigate Colitis in Mice via Inhibition of the NLRP3 Signaling Pathway and Modulation of the Gut Microbiota. Int J Nanomedicine 2024; 19:13991-14018. [PMID: 39742094 PMCID: PMC11687308 DOI: 10.2147/ijn.s493434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Plant-derived exosome-like nanoparticles (PELNs) have received widespread attention in treating ulcerative colitis (UC). However, the role of Houttuynia cordata-derived exosome-like nanoparticles (HELNs) in UC remains unclear. This study aims to evaluate the efficacy of HELNs in treating colitis in mice and investigate its potential mechanisms. METHODS HELNs were isolated from H. cordata for characterization, and their safety and stability were evaluated. A dextran sulfate sodium (DSS)-induced colitis mouse model was utilized to assess the therapeutic potential of HELNs in UC. In vivo, imaging and flow cytometry were utilized to investigate the targeting effect of HELNs on inflamed colonic sites and their modulation of the immune environment. RNA-seq analysis and molecular docking were performed to identify potential pathways recruited by HELNs. Guided by transcriptomic findings, NLRP3-/- mice were used in conjunction with Western blotting, qPCR, immunofluorescence, and other techniques to verify that HELNs alleviated DSS-induced colitis by inhibiting NLRP3/NOD-like receptor signaling pathways. Lastly, the impact of HELNs on the gut microbiota was investigated through 16S rRNA sequencing. RESULTS HELNs significantly reduced the severity of DSS-induced colitis in mice, alleviating colitis symptoms and histopathological damage. Furthermore, HELNs can specifically target inflamed colon tissue, regulate the immune environment, and decrease inflammation. RNA-seq analysis, coupled with the use of NLRP3-/- mice, demonstrated that HELNs inhibited the NLRP3/NOD-like receptor signaling pathways. Lastly, HELNs balanced the gut microbiota composition in mice with colitis, decreasing the abundance of harmful bacteria and increasing the abundance of beneficial bacteria in the intestinal tract of these mice. CONCLUSION In summary, HELNs exhibit the potential to protect the colon from DSS-induced damage by inhibiting the NLRP3/NOD-like receptor signaling pathway and modulating the gut microbiota, presenting a promising therapeutic option for the management of UC.
Collapse
Affiliation(s)
- Jian-Hong Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jin-Xia Hu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yan Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jing-Kui Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yao Peng
- Department of Gastroenterology and Hepatology, Shenzhen General Hospital, Shenzhen, People’s Republic of China
| | - Yong Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - You-Lian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
26
|
Di SJ, Cui XW, Liu TJ, Shi YY. Therapeutic potential of human breast milk-derived exosomes in necrotizing enterocolitis. Mol Med 2024; 30:243. [PMID: 39701931 DOI: 10.1186/s10020-024-01010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 11/23/2024] [Indexed: 12/21/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe inflammatory and necrotizing disease of the intestine that primarily affects the neonates, particularly premature infants. It has a high incidence of approximately 8.9% in extremely preterm infants, with a mortality rate ranging from 20 to 30%. In recent years, exosomes, particularly those derived from breast milk, have emerged as potential candidates for NEC therapy. Human breast milk-derived exosomes (BME) have been shown to enhance intestinal barrier function, protect intestinal epithelial cells from oxidative stress, promote the proliferation and migration of intestinal epithelial cells, and reduce the severity of experimental NEC models. As a subset of extracellular vesicles, BME possess the membrane structure, low immunogenicity, and high permeability, making them ideal vehicles for the treatment of NEC. Additionally, exosomes derived from various sources, including stem cells, intestinal epithelial cells, plants, and bacteria, have been implicated in the development and protection of intestinal diseases. This article summarizes the mechanisms through which exosomes, particularly BME, exert their effects on NEC and discusses the feasibility and obstacles associated with this novel therapeutic strategy.
Collapse
Affiliation(s)
- Si-Jia Di
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xue-Wei Cui
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Tian-Jing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Yong-Yan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
27
|
Liu H, Li M, Xiang B, Yang Z, Cao S, Gong W, Li J, Zhou W, Ding L, Tang Q, Wang S, Tang J, Fan Z, He K, Jiang X, Shen Z, Chen W, Hui J. An integrated "Engage & Evasion" approach for mononuclear phagocyte system escape and efficient extracellular vesicle therapy. J Nanobiotechnology 2024; 22:770. [PMID: 39696354 DOI: 10.1186/s12951-024-03032-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Ischemic diseases are major contributors to global morbidity and mortality, posing a substantial threat to human health. Extracellular vesicles (EVs) play an essential role in enhancing neovascularization in ischemic tissues, thereby facilitating tissue repair and regeneration. However, the utilization of EVs is hindered by their rapid uptake and clearance by the mononuclear phagocyte system (MPS), which markedly impedes their therapeutic efficacy and organ-specific accumulation. Notably, CD47, upon binding to signal regulatory protein alpha, initiates a "don't eat me" signal, enabling immune evasion from the MPS. Our research has demonstrated that phagocytes predominantly engulf CD47low dendritic DC2.4 cell-derived EVs (DV), while engineered CD47high EVs (MV47) experience minimal ingestion. Leveraging these findings, we have developed a dual-faceted "Engage & Evasion" strategy. Initially, DVs were employed to saturate the MPS, serving as the "engage" component. Subsequently, MV47, fortified with CD47, was introduced for "evasion" purposes. This approach effectively minimized entrapment by the liver and spleen, boosted serum concentration, and enhanced final accumulation in non-MPS organs. In summary, our "Engage & Evasion" therapeutic strategy offers a promising avenue to enhance EV therapeutic potential against ischemic challenges through improved systemic distribution.
Collapse
Affiliation(s)
- Hongman Liu
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Cardiovascular Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Mengting Li
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Bing Xiang
- Department of Cardiovascular Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Ziying Yang
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shiyu Cao
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Wen Gong
- Department of Cardiovascular Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Jingjing Li
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Wenjing Zhou
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Liang Ding
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Qingsong Tang
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shengnan Wang
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China
| | - Jin Tang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zixuan Fan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Ke He
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Xuan Jiang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Sun Yat-Sen University, Shenzhen, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Weiqian Chen
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Jie Hui
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, China.
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Dong K, Wang X, Zhou ZJ, Zheng XR, Chang ZP, Zhao R, Liu JJ, Hou RG, Zhang X, Shao YY. Oral Targeted Delivery of Codonopsis Radix Polysaccharide via Succinyl -DHA Functionalized Nanoparticles Efficiently Alleviates Ulcerative Colitis. Int J Nanomedicine 2024; 19:13235-13251. [PMID: 39679251 PMCID: PMC11645468 DOI: 10.2147/ijn.s484575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Ulcerative colitis (UC) is a chronic intestinal disease characterized by spleen-lung qi deficiency and dampness-pathogenic obstruction. Although there are various treatment options available, patients frequently encounter significant drug-related side effects. Previous studies have shown the potential of Codonopsis Radix polysaccharides A (CPA) in treating UC, but their limited bioavailability has restricted their clinical use. Therefore, the objective of this study was to develop a novel formulation that can address the aforementioned limitations and assess its potential advantages. Methods and Results We synthesized a negatively charged amphipathic prodrug called CPA-SA-DHA (CSD), which consists of CPA as the hydrophilic component, and succinic anhydride and docosahexaenoic acid as the hydrophobic segments. The CSD nanoparticles obtained had a particle size of 180.0 ± 3.2 nm, a negative zeta potential of -29.8 ± 5.3 mV, and a uniform shape with a PDI index of 0.230 ± 0.003. The interaction between positive and negative charges significantly increased the retention time of CSD nanoparticles in the colonic microenvironment. Furthermore, CSD nanoparticles demonstrated enhanced bioavailability in UC mice compared to CPA. Additionally, we observed that CSD nanoparticles exhibited therapeutic effects on DSS-induced UC mice by regulating the diversity and abundance of gut microbiota. This effect may be mediated by the inhibition of pro-inflammatory signaling pathways TLR4/NF-κB. Conclusion These findings confirm the potential of CSD nanoparticles as a promising treatment option for UC.
Collapse
Affiliation(s)
- Kang Dong
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xin Wang
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Ze-jia Zhou
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xin-ru Zheng
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Zhuang-peng Chang
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Rui Zhao
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jun-jin Liu
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Rui-gang Hou
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xiao Zhang
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yun-yun Shao
- Department of Pharmacy, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
29
|
Zheng M, Chavda VP, Vaghela DA, Bezbaruah R, Gogoi NR, Patel K, Kulkarni M, Shen B, Singla RK. Plant-derived exosomes in therapeutic nanomedicine, paving the path toward precision medicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156087. [PMID: 39388922 DOI: 10.1016/j.phymed.2024.156087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Plant-derived exosomes (PDEs), are nanoscale vesicles secreted by multivesicular bodies, play pivotal roles in critical biological processes, including gene regulation, cell communication, and immune defense against pathogens. Recognized for their potential health-promoting properties, PDEs are emerging as innovative components in functional nutrition, poised to enhance dietary health benefits. PURPOSE To describe the efficacy of PDEs in nanoform and their application as precision therapy in many disorders. STUDY DESIGN The design of this review was carried out in PICO format using randomized clinical trials and research articles based on in vivo and in vitro studies. METHODS All the relevant clinical and research studies conducted on plant-derived nanovesicle application and efficacy were included, as retrieved from PubMed and Cochrane, after using specific search terms. This review was performed to determine PDEs' efficacy as nanomedicine and precision therapy. Sub-group analysis and primary data were included to determine the relationship with PDEs. RESULT PDEs are extracted from plant materials using sophisticated techniques like precipitation, size exclusion, immunoaffinity capture, and ultracentrifugation, encapsulating vital molecules such as lipids, proteins, and predominantly microRNAs. Although their nutritional impact may be minimal in small quantities, the broader application of PDEs in biomedicine, particularly as vehicles for drug delivery, underscores their significance. They offer a promising strategy to improve the bioavailability and efficacy of therapeutic agents carrying nano-bioactive substances that exhibit anti-inflammatory, antioxidant, cardioprotective, and anti-cancer activities. CONCLUSION PDEs enhance the therapeutic potency of plant-derived phytochemicals, supporting their use in disease prevention and therapy. This comprehensive review explores the multifaceted aspects of PDEs, including their isolation methods, biochemical composition, health implications, and potential to advance medical and nutritional interventions.
Collapse
Affiliation(s)
- Min Zheng
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610218, China
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M College of Pharmacy, Ahmedabad 380009, Gujrat, India.
| | - Dixa A Vaghela
- Pharmacy section, L.M College of Pharmacy Ahmedabad 380009, Gujrat, India
| | - Rajashri Bezbaruah
- Department of Pharmacology, Dibrugarh University, Dibrugarh 786004, Assam
| | - Niva Rani Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh 786004, Assam
| | - Kaushika Patel
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, LJ University, Ahmedabad 382210, Gujarat, India
| | - Mangesh Kulkarni
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, LJ University, Ahmedabad 382210, Gujarat, India; Department of Pharmaceutics, Gandhinagar Institute of Pharmacy, Gandhinagar University, Moti Bhoyan, Khatraj-Kalol Road 382721, Gujarat, India
| | - Bairong Shen
- Institutes for Systems Genetics, West China Tianfu Hospital, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rajeev K Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| |
Collapse
|
30
|
Lv L, Li Z, Liu X, Zhang W, Zhang Y, Liang Y, Zhang Z, Li Y, Ding M, Li R, Lin J. Revolutionizing medicine: Harnessing plant-derived vesicles for therapy and drug transport. Heliyon 2024; 10:e40127. [PMID: 39634409 PMCID: PMC11615498 DOI: 10.1016/j.heliyon.2024.e40127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/29/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
The emergence of extracellular vesicles (EVs), which are natural lipid bilayer membrane structures facilitating intercellular substance and information exchange, has sparked innovative approaches in drug development and carrier enhancement. Plant-derived EVs notably offer advantages including low preparation cost, low immunogenicity, flexible drug delivery, high stability, good tissue permeability, and high inherent medicinal value compared to their animal-derived counterparts. Despite these promising attributes, the research on plant-derived EVs remains fragmented and lacks comprehensive synthesis. This review aims to address this gap by summarizing the isolation methods, biological characteristics, and storage techniques of plant-derived EVs. Additionally, we explore the potential of plant-derived EVs as therapeutic agents and drug carriers for treating various diseases. Finally, we delineate the current impediments to plant-derived EV development and highlight future research directions. By providing a detailed overview, we hope to facilitate further research and application in this emerging field.
Collapse
Affiliation(s)
- Li Lv
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Zhenkun Li
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Xin Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Wenhui Zhang
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Yi Zhang
- Department of Thyroid - Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Ying Liang
- Department of Thyroid - Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Zhixian Zhang
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Yueqiao Li
- Department of Medical Oncology, Yanjin Country People's Hospital, No. 87, Pingjie Street, Yanjin County, Zhaotong, 657500, Yunnan, China
| | - Mingxia Ding
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| | - Rongqing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, Yunnan, China
| | - Jie Lin
- Department of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650101, Yunnan, China
| |
Collapse
|
31
|
Lu Y, Lu D, Li C, Chen L. Exploring Immune Cell Infiltration and Small Molecule Compounds for Ulcerative Colitis Treatment. Genes (Basel) 2024; 15:1548. [PMID: 39766817 PMCID: PMC11728156 DOI: 10.3390/genes15121548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) with a relapsing nature and complex etiology. Bioinformatics analysis has been widely applied to investigate various diseases. This study aimed to identify crucial differentially expressed genes (DEGs) and explore potential therapeutic agents for UC. METHODS The GSE47908 and GSE55306 colon tissue transcriptome gene datasets were downloaded from the Gene Expression Omnibus-NCBI (GEO) database. GEO2R and Gene Set Enrichment Analysis (GSEA) were used to screen for DEGs in patients with UC compared to the normal population based on weighted gene co-expression network analysis (WGCNA). GO-BP analysis and KEGG enrichment analysis were performed on the intersecting differential genes via the Metascape website, while hub genes were analyzed by STRING11.0 and Cytoscape3.7.1. The expression of hub genes was verified in the dataset GSE38713 colon tissue specimens. Finally, the gene expression profiles of the validation set were analyzed by immuno-infiltration through the ImmuCellAI online tool, and the CMap database was used to screen for negatively correlated small molecule compounds. RESULTS A total of 595 and 926 genes were screened by analysis of GSE47908 and GSE55306 datasets, respectively. Combined WGCNA hub module intersection yielded 12 hub genes (CXCL8, IL1β, CXCL1, CCL20, CXCL2, CXCR2, LCN2, SELL, AGT, LILRB3, MMP3, IDO1) associated with the pathogenesis of UC. GSEA analysis yielded intersecting pathways for both datasets (colorectal cancer pathway, base excision repair, cell cycle, apoptosis). GO-BP and KEGG enrichment analyses were performed to obtain key biological processes (inflammatory response, response to bacteria, leukocyte activation involved in the immune response, leukocyte-cell adhesion, apoptosis, positive regulation of immune effector processes) and key signaling pathways (cytokine-cytokine receptor interactions, IBD, NOD-like receptor signaling pathways). The immune cell infiltration analysis suggested that the incidence of UC was mainly related to the increase in CD4+T cells, depletion of T cells, T follicular helper cells, natural killer cells, γδ T cells and the decrease in CD8 naive T cells, helper T cells 17 and effector T cells. The CMap database results showed that small molecule compounds such as vorinostat, roxarsone, and wortmannin may be therapeutic candidates for UC. CONCLUSIONS This study not only aids in early prediction and prevention but also provides novel insights into the pathogenesis and treatment of UC.
Collapse
Affiliation(s)
- Yi Lu
- Shanghai Tufeng Pharmaceutical Technology Co., Ltd., Shanghai 201203, China
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China
| | - Dongqing Lu
- Department of Traditional Chinese Medicine, Beicai Community Health Service Center of Pudong New District, 271 Lianyuan Road, Pudong New District, Shanghai 201024, China
| | - Chujie Li
- Department of Pharmacology and Personalized Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- The M-Lab., Department of Precision Medicine, GROW—Research Institute for Oncology and Repro-Duction, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Luping Chen
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
32
|
Zhang J, Tian S, Guo L, Zhao H, Mao Z, Miao M. Chinese herbal medicine-derived extracellular vesicles as novel biotherapeutic tools: present and future. J Transl Med 2024; 22:1059. [PMID: 39587576 PMCID: PMC11587639 DOI: 10.1186/s12967-024-05892-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
Extracellular vesicles (EVs) are phospholipid bilayer-enclosed biological particles that are secreted by almost all living cells including animals, plants, and microorganisms. Chinese herbal medicines (CHM) have a long history of using plant-based remedies to treat and prevent human diseases. Chinese herbal medicine-derived extracellular vesicle (CHMEV) generic term refers to nanoscale membrane structures isolated from medicinal plants such as ginseng, ginger, and Panax notoginseng. In recent years, CHMEVs have garnered substantial attention as a novel class of functional components due to their high bioavailability, safety, easy accessibility, and diverse therapeutic effects, indicating their great potential for development as a new dosage form of CHM. Research on CHMEVs in traditional Chinese medicine (TCM) has become a prominent area of interest, opening new avenues for further exploration into the therapeutic effects and functional mechanisms of CHM. Nonetheless, as an emerging field, there is much unknown about these vesicles, and current research remains inconsistent. The review comprehensively summarizes the biogenesis, isolation methods, and physical, and biochemical characterizations of CHMEVs. Additionally, we highlight their biomedical applications as therapeutic agents and drug delivery carriers, including anti-inflammatory, anticancer, regenerative, and antiaging activities. Finally, we propose current challenges and future perspectives. By summarizing the existing literature, we aim to offer valuable clues and inspiration for future CHMEV research, thereby facilitating research standardization of CHMEVs in the treatment of human diseases and drug discovery.
Collapse
Affiliation(s)
- Jinying Zhang
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
| | - Shuo Tian
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu- Yao Affiliated to Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
| | - Lin Guo
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
| | - Hui Zhao
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
| | - Zhiguo Mao
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China
| | - Mingsan Miao
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China.
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu- Yao Affiliated to Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, PR China.
| |
Collapse
|
33
|
Zeng YB, Deng X, Shen LS, Yang Y, Zhou X, Ye L, Chen S, Yang DJ, Chen GQ. Advances in plant-derived extracellular vesicles: isolation, composition, and biological functions. Food Funct 2024; 15:11319-11341. [PMID: 39523827 DOI: 10.1039/d4fo04321a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Plant-derived extracellular vesicles (PDEVs) are nanoscale vesicles released from plant cells into the extracellular space. While similar in structure and function to mammalian-derived EVs, PDEVs are unique due to their origin and the specific metabolites they carry. PDEVs have gained significant attention in recent years, with numerous reports isolating different PDEVs from various plants, each exhibiting diverse biological functions. However, the field is still in its early stages, and many issues need further exploration. To better develop and utilize PDEVs, it is essential to have a comprehensive understanding of their characteristics. This review provides an overview of recent advances in PDEV research. It focuses on the methods and techniques for isolating and purifying PDEVs, comparing their respective advantages, limitations, and application scenarios. Furthermore, we discuss the latest discoveries regarding the composition of PDEVs, including lipids, proteins, nucleic acids, and various plant metabolites. Additionally, we detail advanced studies on the multiple biological functions of PDEVs. Our goal is to advance our understanding of PDEVs and encourage further exploration in PDEV-based science and technology, offering insights into their potential applications for human health.
Collapse
Affiliation(s)
- Yao-Bo Zeng
- Department of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 402760, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xun Deng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China.
| | - Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China.
- Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Yong Yang
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China.
- Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Xing Zhou
- Department of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 402760, China
| | - Lianbao Ye
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., China
| | - Da-Jian Yang
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China.
- Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Chongqing 400065, China
| | - Guo-Qing Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong S.A.R., China
| |
Collapse
|
34
|
Zhao Z, Lacombe J, Simon L, Sanchez-Ballester NM, Khanishayan A, Shaik N, Case K, Dugas PY, Repellin M, Lollo G, Soulairol I, Harris AF, Gordon M, Begu S, Zenhausern F. Physical, biochemical, and biological characterization of olive-derived lipid nanovesicles for drug delivery applications. J Nanobiotechnology 2024; 22:720. [PMID: 39558361 PMCID: PMC11575425 DOI: 10.1186/s12951-024-02964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/31/2024] [Indexed: 11/20/2024] Open
Abstract
Extracellular vesicles (EVs) have shown great promise as drug delivery system (DDS). However, their complex and costly production limit their development for clinical use. Interestingly, the plant kingdom can also produce EV-like nanovesicles that can easily be isolated and purified from a large quantity of raw material at a high yield. In this study, olive-derived nanovesicles (ODNVs) were isolated from raw fruits using serial centrifugations and their physical and biological features characterized to demonstrate their promising potential to be used as a DDS. Nanotracking particle analysis indicated an average size of 109.5 ± 3.0 nm and yield of 1012 ODNVs/mL for the purest fraction. Microscopy imaging, membrane fluidity assay and lipidomics analysis showed the presence of a rich lipid bilayer that significantly varied between different sources of ODNVs but showed a distinct signature compared to human EVs. Moreover, ODNVs were enriched in PEN1 and TET8 compared to raw fruits, suggesting an extracellular origin. Interestingly, ODNVs size and yield stayed unchanged after exposure to high temperature (70 °C for 1 h), wide pH range (5-10), and 50-100 nm extrusion, demonstrating high resistance to physical and chemical stresses. This high resistance allowed ODNVs to stay stable in water at 4 °C for a month, or with the addition of 25 mM trehalose for long-term freezing storage. Finally, ODNVs were internalized by both 2D and 3D cell culture without triggering significant cytotoxicity and immunogenicity. Importantly, the anticancer drug doxorubicin (dox) could be loaded by passive incubation within ODNVs and dox-loaded ODNVs decreased cell viability by 90% compared to only 70% for free dox at the same concentration, indicating a higher efficiency of drug delivery by ODNVs. In addition, this high cytotoxicity effect of dox-loaded ODNVs was shown to be stable after a 2-week storage at 4 °C. Together, these findings suggested that ODNVs represent a promising candidate as drug nanocarrier for various DDS clinical applications, as demonstrated by their biocompatibility, high resistance to stress, good stability in harsh environment, and improvement of anticancer drug efficacy.
Collapse
Affiliation(s)
- Zhu Zhao
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA.
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA.
| | - Laurianne Simon
- ICGM, CNRS, ENSCM, University of Montpellier, Montpellier, 34000, France
| | - Noelia M Sanchez-Ballester
- ICGM, CNRS, ENSCM, University of Montpellier, Montpellier, 34000, France
- Department of Pharmacy, Nîmes University Hospital, Nîmes, 30900, France
| | - Ashkan Khanishayan
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA
| | - Naina Shaik
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA
| | - Kallie Case
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA
| | - Pierre-Yves Dugas
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, CP2M UMR 5128, Villeurbanne, France
| | - Mathieu Repellin
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 bd 11 Novembre 1918, Villeurbanne, 69622, France
| | - Giovanna Lollo
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 bd 11 Novembre 1918, Villeurbanne, 69622, France
- Institut universitaire de France (IUF), Paris, France
| | - Ian Soulairol
- ICGM, CNRS, ENSCM, University of Montpellier, Montpellier, 34000, France
- Department of Pharmacy, Nîmes University Hospital, Nîmes, 30900, France
| | - Ashlee F Harris
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA
| | - Michael Gordon
- HonorHealth Research Institute, Scottsdale, AZ, 85258, USA
| | - Sylvie Begu
- ICGM, CNRS, ENSCM, University of Montpellier, Montpellier, 34000, France
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA.
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85004, USA.
- HonorHealth Research Institute, Scottsdale, AZ, 85258, USA.
| |
Collapse
|
35
|
Xavier LEMDS, Reis TCG, Martins ASDP, Santos JCDF, Bueno NB, Goulart MOF, Moura FA. Antioxidant Therapy in Inflammatory Bowel Diseases: How Far Have We Come and How Close Are We? Antioxidants (Basel) 2024; 13:1369. [PMID: 39594511 PMCID: PMC11590966 DOI: 10.3390/antiox13111369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Inflammatory bowel diseases (IBD) pose a growing public health challenge with unclear etiology and limited efficacy of traditional pharmacological treatments. Alternative therapies, particularly antioxidants, have gained scientific interest. This systematic review analyzed studies from MEDLINE, Cochrane, Web of Science, EMBASE, and Scopus using keywords like "Inflammatory Bowel Diseases" and "Antioxidants." Initially, 925 publications were identified, and after applying inclusion/exclusion criteria-covering studies from July 2015 to June 2024 using murine models or clinical trials in humans and evaluating natural or synthetic substances affecting oxidative stress markers-368 articles were included. This comprised 344 animal studies and 24 human studies. The most investigated antioxidants were polyphenols and active compounds from medicinal plants (n = 242; 70.3%). The review found a strong link between oxidative stress and inflammation in IBD, especially in studies on nuclear factor kappa B and nuclear factor erythroid 2-related factor 2 pathways. However, it remains unclear whether inflammation or oxidative stress occurs first in IBD. Lipid peroxidation was the most studied oxidative damage, followed by DNA damage. Protein damage was rarely investigated. The relationship between antioxidants and the gut microbiota was examined in 103 animal studies. Human studies evaluating oxidative stress markers were scarce, reflecting a major research gap in IBD treatment. PROSPERO registration: CDR42022335357 and CRD42022304540.
Collapse
Affiliation(s)
| | | | - Amylly Sanuelly da Paz Martins
- Postgraduate Studies at the Northeast Biotechnology Network (RENORBIO), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| | - Juliana Célia de Farias Santos
- Postgraduate Degree in Medical Sciences (PPGCM/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| | - Nassib Bezerra Bueno
- Postgraduate Degree in Nutrition (PPGNUT), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil; (L.E.M.d.S.X.); (N.B.B.)
| | - Marília Oliveira Fonseca Goulart
- Postgraduate Studies at the Northeast Biotechnology Network (RENORBIO), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
- Institute of Chemistry and Biotechnology (IQB/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil
| | - Fabiana Andréa Moura
- Postgraduate Degree in Nutrition (PPGNUT), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil; (L.E.M.d.S.X.); (N.B.B.)
- Postgraduate Degree in Medical Sciences (PPGCM/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| |
Collapse
|
36
|
Zhao J, Jia W, Zhang R, Wang X, Zhang L. Improving curcumin bioavailability: Targeted delivery of curcumin and loading systems in intestinal inflammation. Food Res Int 2024; 196:115079. [PMID: 39614566 DOI: 10.1016/j.foodres.2024.115079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 12/01/2024]
Abstract
Curcumin is a natural food ingredient and has the potential to alleviate inflammation and combat cancer. The incidence of intestinal inflammation has been increasing and poses a severe risk to human health. Due to low absorption and bioavailability, curcumin's anti-inflammatory ability is ineffective. To improve the bioavailability of curcumin, descriptions of the intestinal barrier, signaling pathways, and transport mechanisms are reviewed. Blocking the signaling pathways lowers the number of inflammatory cytokines produced, which is the primary mechanism by which curcumin relieves inflammatory symptoms. The bioavailability of curcumin is not only related to physicochemical properties but also to the nature of the carrier material. Environmental indicators also have an impact on the improvement of curcumin bioavailability in applications. There is a need to develop multifunctional and more stable nanomaterial targeting systems to improve curcumin bioavailability and achieve better results in nanotechnology research and targeted inflammation therapy.
Collapse
Affiliation(s)
- Junyi Zhao
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China.
| | - Rong Zhang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Xin Wang
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Li Zhang
- Keyi Sunshine Test, Xi'an 710021, China
| |
Collapse
|
37
|
Ran K, Wang J, Li D, Jiang Z, Ding B, Yu F, Hu S, Wang L, Sun W, Xu H. Sustained-release of SOD from multivesicular liposomes accelerated the colonic mucosal healing of colitis mice by inhibiting oxidative stress. Colloids Surf B Biointerfaces 2024; 243:114143. [PMID: 39128435 DOI: 10.1016/j.colsurfb.2024.114143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/19/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
Oxidative stress has long been known as a pathogenic factor of ulcerative colitis. Superoxide dismutase (SOD) has been demonstrated to mitigate gut mucosal injury via combating oxidative stress. Herein, we developed SOD-loaded multivesicular liposomes (S-MVLs) as sustained-release depot for ulcerative colitis treatment. S-MVLs were spherical honeycomb-like particles with average particle size of 27.3 ± 5.4 μm and encapsulating efficiency of 78.7 ± 2.6 %. Moreover, the two-phase release profiles of SOD from S-MVLs were exhibited, that was, the burst release phase within 4 h and the sustained-release phase within 96 h. After intraperitoneally injecting S-MVLs, in situ retention time of SOD at bowel cavity extended by 4-fold in comparison with SOD solution. In vitro cells experiment showed that S-MVLs had the protective effect on LPS-treated RAW 264.7 cells via scavenging ROS and inhibiting pro-inflammatory cytokines production. S-MVLs ameliorated the body weight loss, DAI score and the colon shortening of colitis mice. Meanwhile, the colonic morphology and the epithelial barrier of colitis mice were effectively recovered after S-MVLs treatment. The therapeutic mechanism might be associated with polymerizing M1 macrophages to M2 phenotypes and alleviating oxidative stress. Collectively, multivesicular liposomes might be a promising sustained-release depot of SOD for ulcerative colitis treatments.
Collapse
Affiliation(s)
- Kunjie Ran
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jie Wang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Dingwei Li
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Zhijiang Jiang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Bingyu Ding
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Fengnan Yu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| | - Lifen Wang
- Research Center for Drug Safety Evaluation, Hainan Medical University, Haikou City, Hainan Province, China.
| | - Wenwen Sun
- Pathology Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| | - Helin Xu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China.
| |
Collapse
|
38
|
Zhang S, Li G, Qian K, Zou Y, Zheng X, Ai H, Lin F, Lei C, Hu S. Exosomes derived from cancer cells relieve inflammatory bowel disease in mice. J Drug Target 2024; 32:1073-1085. [PMID: 38958251 DOI: 10.1080/1061186x.2024.2369876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Exosome therapy has garnered significant attention due to its natural delivery capabilities, low toxicity, high biocompatibility, and potential for personalised treatment through engineering modifications. Recent studies have highlighted the ability of tumour cell-derived exosomes (TDEs) to interact with immune cells or modify the immune microenvironment to suppress host immune responses, as well as their unique homing ability to parental cells. The core question of this study is whether this immunomodulatory property of TDEs can be utilised for the immunotherapy of inflammatory diseases. In our experiments, we prepared exosomes derived from murine colon cancer cells CT26 (CT26 exo) using ultracentrifugation, characterised them, and conducted proteomic analysis. The therapeutic potential of CT26 exo was evaluated in our dextran sulphate sodium salt (DSS)-induced inflammatory bowel disease (IBD) mouse model. Compared to the control and 293 T exo treatment groups, mice treated with CT26 exo showed a reduction in the disease activity index (DAI) and colon shortening rate, with no noticeable weight loss. Haematoxylin and eosin (H&E) staining of colon paraffin sections revealed reduced inflammatory infiltration and increased epithelial goblet cells in the colons of CT26 exo-treated group. Furthermore, we conducted preliminary mechanistic explorations by examining the phenotyping and function of CD4+ T cells and dendritic cells (DCs) in the colonic lamina propria of mice. The results indicated that the ameliorative effect of CT26 exosomes might be due to their inhibition of pro-inflammatory cytokine secretion by colonic DCs and selective suppression of Th17 cell differentiation in the colon. Additionally, CT26 exo exhibited good biosafety. Our findings propose a novel exosome-based therapeutic approach for IBD and suggest the potential application of TDEs in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Shuyi Zhang
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Guangyao Li
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Kewen Qian
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Yitan Zou
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xinya Zheng
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Hongru Ai
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Fangxing Lin
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Shi Hu
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| |
Collapse
|
39
|
Zheng M, Song W, Huang P, Huang Y, Lin H, Zhang M, He H, Wu J. Drug conjugates crosslinked bioresponsive hydrogel for combination therapy of diabetic wound. J Control Release 2024; 376:701-716. [PMID: 39447843 DOI: 10.1016/j.jconrel.2024.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Basic fibroblast growth factor (bFGF) has proved to be effective for wound healing, yet its effectiveness is extremely retarded in diabetic wounds due to the severe oxidative stress in wound beds. To solve this issue, herein a novel combination therapy of bFGF and N-acetylcysteine (NAC, antioxidant) was devised for improved diabetic wound repair. To avoid rapid loss of both drugs in the wound beds, a bioresponsive hydrogel (bFGF-HSPP-NAC) was engineered by incorporating bFGF and NAC into polymer-drug conjugates (HSPP) via thiol-disulfide exchange reactions. In response to oxidative stress (e.g., reactive oxygen species), the disulfide bonds (SS) within the hydrogel are broken into thiol groups (-S-H), thereby promoting hydrogel degradation and enabling controlled drug release. Initially, NAC is released to scavenge free radicals and ameliorate oxidative damage. Subsequently, bFGF is released to expedite tissue regeneration. This combinatorial strategy is tailored to the specific characteristics of the wound microenvironment at various stages of diabetic wound healing, thereby achieving therapeutic efficacy. The results indicate that the bFGF-HSPP-NAC hydrogel markedly enhances re-epithelialization, collagen deposition, hair follicle regeneration, and neovascularization. In conclusion, the bioresponsive bFGF-HSPP-NAC hydrogel demonstrates significant potential for application in combinatorial therapeutic approaches for diabetic wound healing.
Collapse
Affiliation(s)
- Manhui Zheng
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, PR China
| | - Wenxiang Song
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang 325035, PR China
| | - Peipei Huang
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang 325035, PR China
| | - Yueping Huang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Hanxuan Lin
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Miao Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, PR China; College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang 325035, PR China.
| | - Jiang Wu
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, PR China.
| |
Collapse
|
40
|
Yang Y, Zhang C, Lin L, Li Q, Wang M, Zhang Y, Yu Y, Hu D, Wang X. Multifunctional MnGA Nanozymes for the Treatment of Ulcerative Colitis by Reducing Intestinal Inflammation and Regulating the Intestinal Flora. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56884-56901. [PMID: 39401179 DOI: 10.1021/acsami.4c14291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
In ulcerative colitis (UC), the formation of an inflammatory environment is due to the combined effects of excess production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), overproduction of proinflammatory cytokines, and disruption of immune system function. There are many kinds of traditional drugs for the clinical treatment of UC, but long-term drug use can cause toxic side effects and drug resistance and can also reduce patient compliance and other drawbacks. Hence, in light of the clinical challenges associated with UC, including the limitations of existing treatments, intense adverse reactions and the development of resistance to medications, no novel therapeutic agents that offer effective relief and maintain a high level of biosafety are urgently needed. Although many anti-inflammatory nanomedicines have been developed by researchers, the development of efficient and nontoxic nanomedicines is still a major challenge in clinical medicine. Using the natural product gallic acid and the metal compound manganese chloride, a highly effective and nontoxic multifunctional nanoenzyme was developed for the treatment of UC. Nanozymes can effectively eliminate ROS and RNS to reduce the inflammation of intestinal epithelial cells caused by oxidation, facilitate the restoration of the intestinal epithelial barrier through the upregulation of tight junction protein expression, and balance the intestinal microbiota to maintain the stability of the intestinal environment. Using a rodent model designed to mimic UC, we monitored body weight, colon length, the spleen index, and the degree of tissue damage and demonstrated that manganese gallate (MnGA) nanoparticles can reduce intestinal inflammation by clearing ROS and active nitrogen. Intestinal flora sequencing revealed that MnGA nanoparticles could regulate the intestinal flora, promote the growth of beneficial bacteria and decrease the levels of detrimental bacteria within the intestinal tract in a mouse model of UC. Thus, MnGA nanoparticles can maintain the balance of the intestinal flora. This study demonstrated that MnGA nanoparticles are excellent antioxidant and effective anti-inflammatory agents, have good biosafety, and can effectively treat UC.
Collapse
Affiliation(s)
- Yan Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Cong Zhang
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Liting Lin
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Qingrong Li
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Min Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Yiqun Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Yue Yu
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Duanmin Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
41
|
Liang Y, Li Y, Lee C, Yu Z, Chen C, Liang C. Ulcerative colitis: molecular insights and intervention therapy. MOLECULAR BIOMEDICINE 2024; 5:42. [PMID: 39384730 PMCID: PMC11464740 DOI: 10.1186/s43556-024-00207-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by abdominal pain, diarrhea, rectal bleeding, and weight loss. The pathogenesis and treatment of UC remain key areas of research interest. Various factors, including genetic predisposition, immune dysregulation, and alterations in the gut microbiota, are believed to contribute to the pathogenesis of UC. Current treatments for UC include 5-aminosalicylic acids, corticosteroids, immunosuppressants, and biologics. However, study reported that the one-year clinical remission rate is only around 40%. It is necessary to prompt the exploration of new treatment modalities. Biologic therapies, such as anti-TNF-α monoclonal antibody and JAK inhibitor, primarily consist of small molecules targeting specific pathways, effectively inducing and maintaining remission. Given the significant role of the gut microbiota, research into intestinal microecologics, such as probiotics and prebiotics, and fecal microbiota transplantation (FMT) shows promising potential in UC treatment. Additionally, medicinal herbs, such as chili pepper and turmeric, used in complementary therapy have shown promising results in UC management. This article reviews recent findings on the mechanisms of UC, including genetic susceptibility, immune cell dynamics and cytokine regulation, and gut microbiota alterations. It also discusses current applications of biologic therapy, herbal therapy, microecologics, and FMT, along with their prospects and challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yang Li
- Department of Respiratory, Sichuan Integrative Medicine Hospital, Chengdu, 610042, China
| | - Chehao Lee
- Department of Traditional Chinese Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziwei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chongli Chen
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chao Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
42
|
Cui C, Du M, Zhao Y, Tang J, Liu M, Min G, Chen R, Zhang Q, Sun Z, Weng H. Functional Ginger-Derived Extracellular Vesicles-Coated ZIF-8 Containing TNF-α siRNA for Ulcerative Colitis Therapy by Modulating Gut Microbiota. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53460-53473. [PMID: 39303016 DOI: 10.1021/acsami.4c10562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Tumor necrosis factor-α (TNF-α) plays a causal role in the pathogenesis of ulcerative colitis (UC), and anti-TNF-α siRNA shows great promise in UC therapy. However, delivering siRNA with site-targeted stability and therapeutic efficacy is still challenging due to the complex and dynamic intestinal microenvironment. Here, based on the functional plant-derived ginger extracellular vesicles (EVs) and porous ZIF-8 nanoparticles, we propose a novel TNF-α siRNA delivery strategy (EVs@ZIF-8@siRNA) for UC targeted therapy. Ginger EVs show strong colon and macrophage targeting, as well as robust resistance to acidic degradation in the stomach. Moreover, 6-shogaol in ginger-derived EVs displays anti-inflammatory effects, which enhance the treatment efficiency by cooperation with TNF-α siRNA. In vitro experiments reveal that ZIF-8 nanoparticles have high TNF-α siRNA loading capacity and promote siRNA escape from cellular lysosomes. In vivo experiments show that the TNF-α level is reduced more significantly in colonic tissue than other nontargeted inflammation related factors, showing a good targeting of this composite nanoparticle. Furthermore, gut microbiota sequencing results demonstrate that the nanoparticles can promote intestinal barrier repair by regulating the intestinal microbial balance and restoring the intestinal health of UC mice. Therefore, the developed EVs@ZIF-8@siRNA nanoparticles may represent a novel colon-targeted oral drug, providing a promising therapeutic strategy for UC therapy.
Collapse
Affiliation(s)
- Chenyang Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Miao Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Yihang Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Jiaze Tang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Mengge Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Geng Min
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Rongchen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Qiang Zhang
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhaowei Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Haibo Weng
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| |
Collapse
|
43
|
Zhang M, Xu X, Su L, Zeng Y, Lin J, Li W, Zou Y, Li S, Lin B, Li Z, Chen H, Huang Y, Xu Q, Chen H, Cheng F, Dai D. Oral administration of Sophora Flavescens-derived exosomes-like nanovesicles carrying CX5461 ameliorates DSS-induced colitis in mice. J Nanobiotechnology 2024; 22:607. [PMID: 39379937 PMCID: PMC11463058 DOI: 10.1186/s12951-024-02856-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
Ulcerative colitis (UC) belongs to chronic inflammatory disease with a relapsing characterization. Conventional oral drugs of UC are restricted in clinical by premature degradation in the gastrointestinal tract, modest efficacy, and adverse effects. CX5461 can treat autoimmune disease, immunological rejection, and vascular inflammation. However, low solubility, intravenous administration, and non-inflammatory targeting limited its clinical application. Herein, this work aims to develop Sophora Flavescens-derived exosomes-like nanovesicles carrying CX5461 (SFELNVs@CX5461) for efficient CX5461 oral delivery for UC therapy. We identified SFELNVs as nano-diameter (80 nm) with negative zeta potential (-32mV). Cellular uptake has shown that SFELNVs were targeted uptake by macrophages, thus increasing drug concentration. Additionally, oral SFELNVs@CX5461 exhibited good safety and stability, as well as inflammation-targeting ability in the gastrointestinal tract of dextran sodium sulfate (DSS)-induced colitis mice. In vivo, oral administration of SFELNVs and CX5461 could relieve mice colitis. More importantly, combined SFELNVs and CX5461 alleviated mice colitis by inhibiting pro-inflammatory factors (TNF-α, IL-1β, and IL-6) expression and promoting M2 macrophage polarization. Furthermore, SFELNVs promoted M2 polarization by miR4371c using miRNA sequencing. Our results suggest that SFELNVs@CX5461 represents a novel orally therapeutic drug that can ameliorate colitis, and a promising targeting strategy for safe UC therapy.
Collapse
Affiliation(s)
- Manqi Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Xichao Xu
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518052, China
| | - Liqian Su
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yuqing Zeng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Jingxiong Lin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Wenwen Li
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
| | - Yigui Zou
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
| | - Sicong Li
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
| | - Boxian Lin
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
| | - Ziyuan Li
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
| | - Hu Chen
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China
| | - Yuheng Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China
| | - Quanle Xu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China.
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Gongchang Road, Shenzhen, Guangdong, 518107, China.
| | - Dongling Dai
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children's Hospital, Shenzhen, 518036, China.
| |
Collapse
|
44
|
D’Angeli F, Granata G, Romano IR, Distefano A, Lo Furno D, Spila A, Leo M, Miele C, Ramadan D, Ferroni P, Li Volti G, Accardo P, Geraci C, Guadagni F, Genovese C. Biocompatible Poly(ε-Caprolactone) Nanocapsules Enhance the Bioavailability, Antibacterial, and Immunomodulatory Activities of Curcumin. Int J Mol Sci 2024; 25:10692. [PMID: 39409022 PMCID: PMC11476408 DOI: 10.3390/ijms251910692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Curcumin (Cur), the primary curcuminoid found in Curcuma longa L., has garnered significant attention for its potential anti-inflammatory and antibacterial properties. However, its hydrophobic nature significantly limits its bioavailability. Additionally, adipose-derived stem cells (ADSCs) possess immunomodulatory properties, making them useful for treating inflammatory and autoimmune conditions. This study aims to verify the efficacy of poly(ε-caprolactone) nanocapsules (NCs) in improving Cur's bioavailability, antibacterial, and immunomodulatory activities. The Cur-loaded nanocapsules (Cur-NCs) were characterized for their physicochemical properties (particle size, polydispersity index, Zeta potential, and encapsulation efficiency) and stability over time. A digestion test simulated the behavior of Cur-NCs in the gastrointestinal tract. Micellar phase analyses evaluated the Cur-NCs' bioaccessibility. The antibacterial activity of free Cur, NCs, and Cur-NCs against various Gram-positive and Gram-negative strains was determined using the microdilution method. ADSC viability, treated with Cur-NCs and Cur-NCs in the presence or absence of lipopolysaccharide, was analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide assay. Additionally, ADSC survival was assessed through the Muse apoptotic assay. The expression of both pro-inflammatory (interleukin-1β and tumor necrosis factor-α) and anti-inflammatory (IL-10 and transforming growth factor-β) cytokines on ADSCs was evaluated by real-time polymerase chain reaction. The results demonstrated high stability post-gastric digestion of Cur-NCs and elevated bioaccessibility of Cur post-intestinal digestion. Moreover, Cur-NCs exhibited antibacterial activity against Escherichia coli without affecting Lactobacillus growth. No significant changes in the viability and survival of ADSCs were observed under the experimental conditions. Finally, Cur-NCs modulated the expression of both pro- and anti-inflammatory cytokines in ADSCs exposed to inflammatory stimuli. Collectively, these findings highlight the potential of Cur-NCs to enhance Cur's bioavailability and therapeutic efficacy, particularly in cell-based treatments for inflammatory diseases and intestinal dysbiosis.
Collapse
Affiliation(s)
- Floriana D’Angeli
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Giuseppe Granata
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123 Catania, Italy; (I.R.R.); (D.L.F.)
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy; (A.D.); (G.L.V.)
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123 Catania, Italy; (I.R.R.); (D.L.F.)
| | - Antonella Spila
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Mariantonietta Leo
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Chiara Miele
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Dania Ramadan
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Patrizia Ferroni
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
- InterInstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele, 00166 Rome, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy; (A.D.); (G.L.V.)
| | - Paolo Accardo
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Corrada Geraci
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Fiorella Guadagni
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
- InterInstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele, 00166 Rome, Italy
| | - Carlo Genovese
- Department of Medicine and Surgery, “Kore” University of Enna, Contrada Santa Panasia, 94100 Enna, Italy;
- Nacture S.r.l, Spin-Off University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
45
|
Zhu Y, Zhao J, Ding H, Qiu M, Xue L, Ge D, Wen G, Ren H, Li P, Wang J. Applications of plant-derived extracellular vesicles in medicine. MedComm (Beijing) 2024; 5:e741. [PMID: 39309692 PMCID: PMC11413507 DOI: 10.1002/mco2.741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Plant-derived extracellular vesicles (EVs) are promising therapeutic agents owing to their natural abundance, accessibility, and unique biological properties. This review provides a comprehensive exploration of the therapeutic potential of plant-derived EVs and emphasizes their anti-inflammatory, antimicrobial, and tumor-inhibitory effects. Here, we discussed the advancements in isolation and purification techniques, such as ultracentrifugation and size-exclusion chromatography, which are critical for maintaining the functional integrity of these nanovesicles. Next, we investigated the diverse administration routes of EVs and carefully weighed their respective advantages and challenges related to bioavailability and patient compliance. Moreover, we elucidated the multifaceted mechanisms of action of plant-derived EVs, including their roles in anti-inflammation, antioxidation, antitumor activity, and modulation of gut microbiota. We also discussed the impact of EVs on specific diseases such as cancer and inflammatory bowel disease, highlighting the importance of addressing current challenges related to production scalability, regulatory compliance, and immunogenicity. Finally, we proposed future research directions for optimizing EV extraction and developing targeted delivery systems. Through these efforts, we envision the seamless integration of plant-derived EVs into mainstream medicine, offering safe and potent therapeutic alternatives across various medical disciplines.
Collapse
Affiliation(s)
- Yawen Zhu
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Junqi Zhao
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Haoran Ding
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Mengdi Qiu
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Lingling Xue
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Dongxue Ge
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Gaolin Wen
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Haozhen Ren
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Peng Li
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
46
|
Sharma V, Sinha ES, Singh J. Investigation of In Vitro Anti-cancer and Apoptotic Potential of Onion-Derived Nanovesicles Against Prostate and Cervical Cancer Cell Lines. Appl Biochem Biotechnol 2024; 196:6957-6973. [PMID: 38441782 DOI: 10.1007/s12010-024-04872-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 11/21/2024]
Abstract
Plant-derived compounds have recently garnered significant interest in the field of medicine due to their rich repertoire of phytochemicals, which holds promise for exploring novel therapies to treat cancer. This study embarks on the first-time investigation of the anti-cancerous effect of onion-derived nanovesicles (ODNVs). ODNVs were isolated employing differential centrifugation followed by ultracentrifugation and subsequent characterization using dynamic light scattering (DLS), field emission scanning electron microscopy (FESEM), and Fourier transform infrared spectroscopy (FTIR). Furthermore, we delineated the anti-cancerous effect of ODNVs on two cancer cell line models HeLa (cervical cancer) and PC-3 (prostate cancer) using MTT assay, DAPI-based DNA damage using immunofluorescence microscopy, colony formation assay, migration assay, cell cycle analysis, and evaluation of apoptosis using flow cytometry and western blotting. The findings revealed dose- and time-dependent anti-proliferative effects of ODNVs on both HeLa and PC3 cell lines, accompanied by selective cytotoxicity against cancer cells. Additional results highlighted that ODNVs prevented colony growth and induced S-phase cell cycle arrest. Apoptosis induction was evaluated through alterations in nuclear morphology and the number of apoptotic cells, which increased significantly after ODNV treatment in both cancer cell lines. Furthermore, annexin V/PI staining evaluation of apoptotic cells by flow cytometry demonstrated that ODNV treatment significantly increased the number of apoptotic cells in both PC-3 and HeLa cells. Finally, Western blot analysis indicated changes in apoptosis-related proteins including bcl-2, bax, and caspase-3, emphasizing that the anti-cancerous effect of ODNVs is attributed to the induction of apoptosis and suggests the unexplored anti-cancerous potential of ODNVs.
Collapse
Affiliation(s)
- Vinayak Sharma
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Eshu Singhal Sinha
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Jagtar Singh
- Department of Biotechnology, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
47
|
Shrestha P, Duwa R, Lee S, Kwon TK, Jeong JH, Yook S. ROS-responsive thioketal nanoparticles delivering system for targeted ulcerative colitis therapy with potent HDAC6 inhibitor, tubastatin A. Eur J Pharm Sci 2024; 201:106856. [PMID: 39032536 DOI: 10.1016/j.ejps.2024.106856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Ulcerative colitis (UC) is a common gastrointestinal problem characterized by the mucosal injury primarily affecting the large intestine. Currently available therapies are not satisfactory as evidenced by high relapse rate and adverse effects. In this study we aimed to develop an effective drug delivery system using reactive oxygen species (ROS)-responsive thioketal nanoparticles (TKNP), to deliver tubastatin A, a potent HDAC6 inhibitor, to the inflamed colon in mice with ulcerative colitis (UC). TKNPs were synthesized by step-growth polymerization from an acetal exchange reaction while TUBA-TKNP was prepared using the single emulsion solvent evaporation technique. Our developed nanoparticle showed release of tubastatin A only in presence of ROS which is found to be highly present at the site of inflamed colon. Oral administration of TUBA-TKNP resulted in the higher accumulation of tubastatin A at the inflamed colon site and decreased the inflammation as evidenced by reduced infiltration of immune cells and decreased level of pro-inflammatory cytokines in TUBA-TKNP treated mice. In summary, our results show the successful localization of tubastatin A at the site of colon inflammation through TUBA-TKNP delivery, as well as resolution of clinical features of UC in mice.
Collapse
Affiliation(s)
- Prabhat Shrestha
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ramesh Duwa
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Radiology, Molecular Imaging Program at Standford (MIPS), School of Medicine, Standford University, Standford, California 94305, USA
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
48
|
Li Z, Du Y, Lu Y, Ma X, Li F, Zeng P, Zhang T, He Y, Luo P, Wu J. Hypericum perforatum-derived exosomes-like nanovesicles for adipose tissue photodynamic therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155854. [PMID: 39032276 DOI: 10.1016/j.phymed.2024.155854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/30/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Recent investigations underscore the capacity of photodynamic therapy (PDT) to induce adipocyte apoptosis, thereby mitigating obesity. Nonetheless, extant synthetic photosensitizers manifest limitations that hinder their clinical viability. PURPOSE In the current study, we used Hypericum perforatum-derived exosomes-like nanovesicles (HPExos) as a novel photosensitizer, and investigated its PDT effects in adipose tissue during obesity. METHOD HPExos-were administered to high fat diet mice via intraperitoneal injection, followed by targeted irradiation with specialized LED lights. Mass spectrometric analysis was analyzed in adipose tissues. CCK8 assay and Oil Red O staining were used to investigate lipid accumulation in 3T3-L1 cells to clarify adipocyte differentiation. The expression levels of related markers associated with adipogenesis and lipogenesis were assessed by RT-PCR. Apoptosis analysis was performed by TUNEL staining of and western blotting. RESULTS HPExos combined with PDT accumulated in visceral white adipose tissues results in a reduced body weight and improved insulin sensitivity. HPExos combined with PDT induced apoptosis by driving high levels of ROS. In addition, HPExos combined with PDT significantly downregulated the expression of transcription factors, PPARγ, C/EBPα, and SREBP and lipogenesis protein FABP4 both in vitro and in vivo, associated with a decreased FFA levels. CONCLUSION These findings suggest that HPExos could act as an effective photosensitizer in regulating glucose hemostasis by inhibiting adipocyte differentiation and lipogenesis, offering a promising approach for obesity treatment.
Collapse
Affiliation(s)
- Ziyu Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, Macau, China; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yu Du
- Department of Rheumatology and Immunology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, China
| | - Yu Lu
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoyu Ma
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fei Li
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Peiyuan Zeng
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tao Zhang
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuqian He
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pei Luo
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China; State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, Macau, China
| | - Jianbo Wu
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
49
|
Wang J, Zhang T, Gu R, Ke Y, Zhang S, Su X, Pan X, He Q, Li G, Zhang Z, Zhang L, Li J, Wu W, Chen C. Development and Evaluation of Reconstructed Nanovesicles from Turmeric for Multifaceted Obesity Intervention. ACS NANO 2024; 18:23117-23135. [PMID: 39153188 DOI: 10.1021/acsnano.4c05309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
The escalating prevalence of obesity poses significant health challenges due to its direct association with various diseases. Most existing medications, such as appetite suppressants and fat absorption inhibitors, suffer from limited effectiveness and undesirable side effects. Here, inspired by the versatile metabolic effects of turmeric, we developed a naturally derived nanoformulation of "Reconstructed Turmeric-derived Nanovesicles (Rec-tNVs)" for obesity treatment. Employing quantitative nanoflow cytometry, a four-orders-of-magnitude increase in curcumin content (∼108 molecules per particle) was identified in individual Rec-tNVs compared to their ultracentrifugation-isolated counterparts. Rec-tNVs, featuring highly aggregated curcumin arrangements and other coencapsulated bioactive compounds, demonstrated a dose-dependent lipid-lowering effect in mature 3T3-L1 cells by promoting lipolysis, suppressing lipogenesis, inducing adipocyte browning, and triggering apoptosis after internalization via multiple pathways. In vivo experiments revealed that Rec-tNVs alleviated obesity more effectively than free curcumin and achieved weight reductions of 18.68 and 14.56% through intragastric and subcutaneous delivery, respectively, in high-fat-diet mouse models over a four-week treatment period. These effects were attributed to targeted actions on adipose tissues and systemic impacts on metabolism and gut microbiota composition. Overall, this study underscores the multifaceted antiobesity efficacy of Rec-tNVs, and offers a promising paradigm for developing plant-derived nanovesicle-based therapeutics.
Collapse
Affiliation(s)
- Jialin Wang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Tianyu Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Ruilan Gu
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yingying Ke
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Siqin Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Xueqi Su
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Xueping Pan
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Qiuxia He
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Guiling Li
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Zhengxiao Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Lingyu Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Jian Li
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Weijing Wu
- Engineering Research Center of Natural Cosmeceuticals College of Fujian Province, Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, Fujian 361018, China
| | - Chaoxiang Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Jimei University, Xiamen, Fujian 361021, China
| |
Collapse
|
50
|
Zeng M, Liu M, Tao X, Yin X, Shen C, Wang X. Emerging Trends in the Application of Extracellular Vesicles as Novel Oral Delivery Vehicles for Therapeutics in Inflammatory Diseases. Int J Nanomedicine 2024; 19:8573-8601. [PMID: 39185348 PMCID: PMC11345024 DOI: 10.2147/ijn.s475532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammation involves complex immune responses where cytokines such as TNF-α, IL-1, and IL-6 promote vasodilation and increased vascular permeability to facilitate immune cell migration to inflammation sites. Persistent inflammation is linked to diseases like cancer, arthritis, and neurodegenerative disorders. Although oral anti-inflammatory drugs are favored for their non-invasiveness and cost-effectiveness, their efficacy is often compromised due to gastrointestinal degradation and limited bioavailability. Recent advancements highlight the potential of extracellular vesicles (EVs) as nanocarriers that enhance drug delivery by encapsulating therapeutic agents, ensuring targeted release and reduced toxicity. These EVs, derived from dietary sources and cell cultures, exhibit excellent biocompatibility and stability, presenting a novel approach in anti-inflammatory therapies. This review discusses the classification and advantages of orally administered EVs (O-EVs), their mechanism of action, and their emerging role in treating inflammatory conditions, positioning them as promising vectors in the development of innovative anti-inflammatory drug delivery systems.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Maozhu Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xuelin Tao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Chao Shen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xueyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|