1
|
Wang Z, Liu Y. Dual-modulation of nutrient-transporter axis and functionalized carriers: A paradigm shift for precision oral vitamin D delivery. Colloids Surf B Biointerfaces 2025; 253:114769. [PMID: 40344743 DOI: 10.1016/j.colsurfb.2025.114769] [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: 03/13/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
The transintestinal epithelial absorption of vitamin D is intricately regulated by specific transport protein networks. Emerging evidence from molecular nutrition research reveals that certain dietary nutrients can enhance intestinal vitamin D absorption through targeted modulation of lipid transport pathways. Despite significant advancements in vitamin D delivery systems demonstrating excellent intestinal mucoadhesion and in vitro bioaccessibility, their clinical translation remains limited by suboptimal in vivo bioavailability. To address this critical challenge, we propose an innovative synergistic nutrient absorption strategy that establishes precise coordination among three key elements: dietary nutrient composition, transport protein regulation, and intestinal absorption optimization. This comprehensive review systematically examines: (1) The molecular mechanisms governing transintestinal vitamin D transport and physiological modulation of protein-mediated absorption pathways; (2) The regulatory effects of dietary components on vitamin D absorption efficiency through protein pathway modulation, proposing a novel "nutrient-transporter-vitamin D axis" strategy integrating cutting-edge carrier technologies; (3) Future perspectives for developing functionalized vitamin D delivery systems. The proposed paradigm shift, combining nutrient-mediated transport enhancement with advanced carrier engineering, represents a transformative approach to overcome current limitations in oral vitamin D delivery. This dual-modulation strategy synergistically improves intestinal absorption and systemic bioavailability through simultaneous optimization of biological transport mechanisms and pharmaceutical delivery parameters, offering new possibilities for precision nutrition interventions.
Collapse
Affiliation(s)
- Zixiao Wang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China.
| |
Collapse
|
2
|
Yu Z, Swift KA, Hedges MA, Theiss AL, Andres SF. Microscopic messengers: Extracellular vesicles shaping gastrointestinal health and disease. Physiol Rep 2025; 13:e70292. [PMID: 40165585 PMCID: PMC11959161 DOI: 10.14814/phy2.70292] [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: 11/25/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The field of extracellular vesicles (EVs) is advancing rapidly, and this review aims to synthesize the latest research connected to EVs and the gastrointestinal tract. We will address new and emerging roles for EVs derived from internal sources such as the pancreas and immune system and how these miniature messengers alter organismal health or the inflammatory response within the GI tract. We will examine what is known about external EVs from dietary and bacterial sources and the immense anti-inflammatory, immune-modulatory, and proliferative potential within these nano-sized information carriers. EV interactions with the intestinal and colonic epithelium and associated immune cells at homeostatic and disease states, such as necrotizing enterocolitis (NEC) and inflammatory bowel disease (IBD) will also be covered. We will discuss how EVs are being leveraged as therapeutics or for drug delivery and conclude with a series of unanswered questions in the field.
Collapse
Affiliation(s)
- Zhantao Yu
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Kevin A. Swift
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Madeline A. Hedges
- Department of Neonatology, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Arianne L. Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
- Rocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
| | - Sarah F. Andres
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| |
Collapse
|
3
|
Longfei H, Wenyuan H, Weihua F, Peng P, Sun L, Kun L, Mincong H, Fan Y, Wei H, Qiushi W. Exosomes in cartilage microenvironment regulation and cartilage repair. Front Cell Dev Biol 2025; 13:1460416. [PMID: 40109360 PMCID: PMC11919854 DOI: 10.3389/fcell.2025.1460416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Osteoarthritis (OA) is a debilitating disease that predominantly impacts the hip, hand, and knee joints. Its pathology is defined by the progressive degradation of articular cartilage, formation of bone spurs, and synovial inflammation, resulting in pain, joint function limitations, and substantial societal and familial burdens. Current treatment strategies primarily target pain alleviation, yet improved interventions addressing the underlying disease pathology are scarce. Recently, exosomes have emerged as a subject of growing interest in OA therapy. Numerous studies have investigated exosomes to offer promising therapeutic approaches for OA through diverse in vivo and in vitro models, elucidating the mechanisms by which exosomes from various cell sources modulate the cartilage microenvironment and promote cartilage repair. Preclinical investigations have demonstrated the regulatory effects of exosomes originating from human cells, including mesenchymal stem cells (MSC), synovial fibroblasts, chondrocytes, macrophages, and exosomes derived from Chinese herbal medicines, on the modulation of the cartilage microenvironment and cartilage repair through diverse signaling pathways. Additionally, therapeutic mechanisms encompass cartilage inflammation, degradation of the cartilage matrix, proliferation and migration of chondrocytes, autophagy, apoptosis, and mitigation of oxidative stress. An increasing number of exosome carrier scaffolds are under development. Our review adopts a multidimensional approach to enhance comprehension of the pivotal therapeutic functions exerted by exosomes sourced from diverse cell types in OA. Ultimately, our aim is to pinpoint therapeutic targets capable of regulating the cartilage microenvironment and facilitating cartilage repair in OA.
Collapse
Affiliation(s)
- Han Longfei
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hou Wenyuan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fang Weihua
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peng Peng
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lu Sun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lin Kun
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Mincong
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yang Fan
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Wei
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Qiushi
- Traumatology and Orthopedics Institute of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Orthopaedic, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Ying Y, Zhang Y, Sun J, Chen Y, Wu H. Mechanism of intestinal flora affecting SLC2A9 transport function to promote the formation of hyperuricemia. Heliyon 2024; 10:e40597. [PMID: 39698087 PMCID: PMC11652827 DOI: 10.1016/j.heliyon.2024.e40597] [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: 09/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
Objective To investigate the structural characteristics of the intestinal flora in obese-hyperuricemic (HUA-W) patients and the mechanisms by which they promote the formation of hyperuricemia. Methods 120 human fecal samples (60 cases in HC, 30 cases in HUA-N, and 30 cases in HUA-W) and 40 cases in the colonic tissues (20 cases in HC, 10 cases in HUA-N, and 10 cases in HUA-W) were collected. The intestinal flora of faeces was detected by 16s rRNA method; and the expression of SLC2A9 on human colon tissues was detected by RT-qPCR method and immunofluorescence method. 40 obese-hyperuricemia rat models were established (10 rats in Model, 10 rats in HC-FT, 10 rats in HUA-N-FT, and 10 rats in HUA-W-FT), and 10 rats were set up in Control; and the level of uric acid in rat serum, the levels of xanthine oxidase (XOD) activity and uric acid in intestinal fluid were examined. SLC2A9+ Caco-2 cells were produced to simulate the Transwell uric acid transport model, and the Caco-2 cells and SLC2A9+ Caco-2 cells were grown in five different culture media (Blank, Germ-free, HC-germ, HUA-N-germ and HUA-W-germ), and the uric acid levels in the upper and lower layers of the chambers were detected. Results The HUA-W intestinal flora showed significant specificity, with a decrease in Bacteroidota and Bacteroidia and an increase in Escherichia and Ruminococcus. There were no significant differences in the fluorescence intensity of the SLC2A9 protein and the SLC2A9 mRNA levels in the colon tissues of the HUA-N and HUA-W (P = 0.447, P = 0.152, P = 0.4799 and P = 0.965, respectively). In rat animal experiments, uric acid levels were significantly higher (P < 0.05) and XOD activity was significantly higher (P < 0.05) in intestinal fluid of HUA-W-FT. In Transwell experiments with SLC2A9+ Caco-2 cells, uric acid levels were increased in the upper compartment and decreased in the lower compartment of HUA-W-germ. Conclusion HUA-W intestinal flora may increase XOD activity in the intestinal tract and improve the ability of uric acid transporter protein SLC2A9 to reabsorb uric acid, providing a new theoretical basis for the pathogenesis of hyperuricemia.
Collapse
Affiliation(s)
- Ying Ying
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Rheumatology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Yi Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Sun
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yong Chen
- Department of Rheumatology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Li Y, Wang Y, Zhao H, Pan Q, Chen G. Engineering Strategies of Plant-Derived Exosome-Like Nanovesicles: Current Knowledge and Future Perspectives. Int J Nanomedicine 2024; 19:12793-12815. [PMID: 39640047 PMCID: PMC11618857 DOI: 10.2147/ijn.s496664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/23/2024] [Indexed: 12/07/2024] Open
Abstract
Plant-derived exosome-like nanovesicles (PELNs) from edible plants, isolated by ultracentrifugation, size exclusion chromatography or other methods, were proved to contain a variety of biologically active and therapeutically specific components. Recently, investigations in the field of PELN-based biomedicine have been conducted, which positioned those nanovesicles as promising tools for prevention and treatment of several diseases, with their natural origin potentially offering superior biocompatibility and bioavailability. However, the inadequate targeting and limited therapeutic effects constrain the utility and clinical translation of PELNs. Thus, strategies aiming at bridging the gap by engineering natural PELNs have been of great interest. Those approaches include membrane hybridization, physical and chemical surface functionalization and encapsulation of therapeutic payloads. Herein, we provide a comprehensive overview of the biogenesis and composition, isolation and purification methods and characterization of PELNs, as well as their therapeutic functions. Current knowledge on the construction strategies and biomedical application of engineered PELNs were reviewed. Additionally, future directions and perspectives in this field were discussed in order to further enrich and expand the prospects for the application of engineered PELNs.
Collapse
Affiliation(s)
- Yuhan Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yulong Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hongrui Zhao
- Intensive Care Medicine Department, Yuhuangding Hospital, Yantai, People’s Republic of China
| | - Qi Pan
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Guihao Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Song Y, Shi M, Wang Y. Deciphering the role of host-gut microbiota crosstalk via diverse sources of extracellular vesicles in colorectal cancer. Mol Med 2024; 30:200. [PMID: 39501131 PMCID: PMC11536884 DOI: 10.1186/s10020-024-00976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/26/2024] [Indexed: 11/09/2024] Open
Abstract
Colorectal cancer is the most common type of cancer in the digestive system and poses a major threat to human health. The gut microbiota has been found to be a key factor influencing the development of colorectal cancer. Extracellular vesicles are important mediators of intercellular communication. Not only do they regulate life activities within the same individual, but they have also been found in recent years to be important mediators of communication between different species, such as the gut microbiota and the host. Their preventive, diagnostic, and therapeutic value in colorectal cancer is being explored. The aim of this review is to provide insights into the complex interactions between host and gut microbiota, particularly those mediated through extracellular vesicles, and how these interactions affect colorectal cancer development. In addition, the potential of extracellular vesicles from various body fluids as biomarkers was evaluated. Finally, we discuss the potential, challenges, and future research directions of extracellular vesicles in their application to colorectal cancer. Overall, extracellular vesicles have great potential for application in medical processes related to colorectal cancer, but their isolation and characterization techniques, intercellular communication mechanisms, and the effectiveness of their clinical application require further research and exploration.
Collapse
Affiliation(s)
- Yun Song
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China.
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, PR China.
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China.
| |
Collapse
|
7
|
Yang L, Liu T, Liao Y, Ren Y, Zheng Z, Zhang M, Yu Y, Liu C, Wang C, Chen T, Zhang L, Zheng D, Zhao H, Ni Z, Liu X. Potential therapeutic application and mechanism of gut microbiota-derived extracellular vesicles in polycystic ovary syndrome. Biomed Pharmacother 2024; 180:117504. [PMID: 39341079 DOI: 10.1016/j.biopha.2024.117504] [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: 07/16/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder affecting women of reproductive age. The syndrome is characterized by androgen excess, ovarian dysfunction, insulin resistance (IR) and obesity, with an elevated risk of developing long-term complications, including cardiovascular disease and type 2 diabetes mellitus (T2D). The gut microbiota plays a role in the pathogenesis of PCOS by influencing the host's endocrine, metabolic and inflammatory state, as well as the gut-brain axis. Gut microbiota-derived extracellular vesicles (GMEVs) are lipid bilayer nanoparticles secreted by the gut microbiota and contain a variety of components, including proteins, lipids and nucleic acids. They serve as signaling molecules, facilitating bacterial-bacterial and bacterial-host communications. Bacterial extracellular vesicles (BEVs) affect host cells through the delivery of bioactive substances and physical interaction through membrane components, thereby participating in the regulation of metabolic, immune, and other cellular processes. Furthermore, BEVs, which are distinguished by low toxicity, high biocompatibility and stability, and the capacity to cross biological barriers, present a promising avenue for the development of novel drug delivery systems. The isolation and characterization of BEVs also facilitate the investigation of disease-specific biomarkers. Consequently, BEVs have immense potential for a range of medical research applications, including disease diagnosis and treatment. This article discusses the potential therapeutic effects and mechanisms of GMEVs in the treatment of PCOS.
Collapse
Affiliation(s)
- Liangliang Yang
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Tingxiu Liu
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yan Liao
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yuehan Ren
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng Zheng
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Mingyue Zhang
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yue Yu
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chang Liu
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Chaoying Wang
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Tong Chen
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Lili Zhang
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Dongxue Zheng
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Haidan Zhao
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Zhexin Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Xinmin Liu
- Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
8
|
Matloob A, Gu X, Rehman Sheikh A, Javed M, Fang Z, Luo Z. Plant exosomes‐like nano‐vesicles: Characterization, functional food potential, and emerging therapeutic applications as a nano medicine. FOOD SAFETY AND HEALTH 2024; 2:429-450. [DOI: 10.1002/fsh3.12060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 01/05/2025]
Abstract
AbstractPlant cells release exosome‐like nanovesicles (PENVs), which are small, membrane‐bound vesicles secreted by cells for intercellular interactions. These vesicles, rich in biologically active substances, are crucial for information transmission, intercellular interaction, and organism homeostasis conservation. They can also be used for treating diseases as large‐scale drug carriers due to their vesicular architecture. This study explores the isolation, potential of nanovesicles in creating bio‐therapeutic and drug‐delivery nano‐platforms to address clinical challenges. The bio‐therapeutic roles of PENVs, include immunomodulation, antitumor, regenerative impacts, wound healing, anti‐fibrosis, whitening effects, and treatment of intestinal flora disorders. This study also deliberates the potential for designing these nanovesicles into effective, safe, and non‐immunogenic nano‐vectors to carry drugs. PENVs may offer a cutting‐edge platform for the creation of functional foods and nutraceuticals. They might be employed to encapsulate certain bioactive substances produced from plants, offering tailored health privileges. It also elucidates the potential for the development of therapeutic and provision nano‐platforms based on PENVs in clinical applications.
Collapse
Affiliation(s)
- Anam Matloob
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Xinya Gu
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Arooj Rehman Sheikh
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Miral Javed
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
| | - Zhongxiang Fang
- School of Agriculture, Food and Ecosystem Sciences Faculty of Science The University of Melbourne Melbourne Victoria Australia
| | - Zisheng Luo
- College of Biosystems Engineering and Food Science Zhejiang University Hangzhou China
- Key Laboratory of Ago‐Products Postharvest Handing of Ministry of Agriculture and Rural Affairs Hangzhou China
| |
Collapse
|
9
|
Cui L, Perini G, Palmieri V, De Spirito M, Papi M. Plant-Derived Extracellular Vesicles as a Novel Frontier in Cancer Therapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1331. [PMID: 39195369 DOI: 10.3390/nano14161331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
Recent advancements in nanomedicine and biotechnology have unveiled the remarkable potential of plant-derived extracellular vesicles (PDEVs) as a novel and promising approach for cancer treatment. These naturally occurring nanoscale particles exhibit exceptional biocompatibility, targeted delivery capabilities, and the capacity to load therapeutic agents, positioning them at the forefront of innovative cancer therapy strategies. PDEVs are distinguished by their unique properties that facilitate tumor targeting and penetration, thereby enhancing the efficacy of drug delivery systems. Their intrinsic biological composition allows for the evasion of the immune response, enabling the efficient transport of loaded therapeutic molecules directly to tumor sites. Moreover, PDEVs possess inherent anti-cancer properties, including the ability to induce cell cycle arrest and promote apoptotic pathways within tumor cells. These vesicles have also demonstrated antimetastatic effects, inhibiting the spread and growth of cancer cells. The multifunctional nature of PDEVs allows for the simultaneous delivery of multiple therapeutic agents, further enhancing their therapeutic potential. Engineering and modification techniques, such as encapsulation, and the loading of therapeutic agents via electroporation, sonication, and incubation, have enabled the customization of PDEVs to improve their targeting efficiency and therapeutic load capacity. This includes surface modifications to increase affinity for specific tumor markers and the encapsulation of various types of therapeutic agents, such as small molecule drugs, nucleic acids, and proteins. Their plant-derived origin offers an abundant and renewable source to produce therapeutic vesicles, reducing costs and facilitating scalability for clinical applications. This review provides an in-depth analysis of the latest research on PDEVs as emerging anti-cancer agents in cancer therapy.
Collapse
Affiliation(s)
- Lishan Cui
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
- Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche CNR, Via dei Taurini 19, 00185 Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| |
Collapse
|
10
|
Liu J, Nordin JZ, McLachlan AJ, Chrzanowski W. Extracellular vesicles as the next-generation modulators of pharmacokinetics and pharmacodynamics of medications and their potential as adjuvant therapeutics. Clin Transl Med 2024; 14:e70002. [PMID: 39167024 PMCID: PMC11337541 DOI: 10.1002/ctm2.70002] [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: 05/20/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND AND MAIN BODY Pharmacokinetics (PK) and pharmacodynamics (PD) are central concepts to guide the dosage and administration of drug therapies and are essential to consider for both healthcare professionals and researchers in therapeutic planning and drug discovery. PK/PD properties of a drug significantly influence variability in response to treatment, including therapeutic failure or excessive medication-related harm. Furthermore, suboptimal PK properties constitute a significant barrier to further development for some candidate treatments in drug discovery. This article describes how extracellular vesicles (EVs) affect different aspects of PK and PD of medications and their potential to modulate PK and PD properties to address problematic PK/PD profiles of drugs. We reviewed EVs' intrinsic effects on cell behaviours and medication responses. We also described how surface and cargo modifications can enhance EV functionalities and enable them as adjuvants to optimise the PK/PD profile of conventional medications. Furthermore, we demonstrated that various bioengineering strategies can be used to modify the properties of EVs, hence enhancing their potential to modulate PK and PD profile of medications. CONCLUSION This review uncovers the critical role of EVs in PK and PD modulation and motivates further research and the development of assays to unfold EVs' full potential in solving PK and PD-related problems. However, while we have shown that EVs play a vital role in modulating PK and PD properties of medications, we postulated that it is essential to define the context of use when designing and utilising EVs in pharmaceutical and medical applications. HIGHLIGHTS Existing solutions for pharmacokinetics and pharmacodynamics modulation are limited. Extracellular vesicles can optimise pharmacokinetics as a drug delivery vehicle. Biogenesis and administration of extracellular vesicles can signal cell response. The pharmaceutical potential of extracellular vesicles can be enhanced by surface and cargo bioengineering. When using extracellular vesicles as modulators of pharmacokinetics and pharmacodynamics, the 'context of use' must be considered.
Collapse
Affiliation(s)
- Jiaqi Liu
- Sydney Pharmacy SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Joel Z. Nordin
- Division of Biomolecular and Cellular MedicineDivision of Clinical ImmunologyDepartment of Laboratory MedicineKarolinska InstituteHuddingeSweden
| | - Andrew J. McLachlan
- Sydney Pharmacy SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Wojciech Chrzanowski
- Sydney Pharmacy SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
- Division of Biomolecular and Cellular MedicineDivision of Clinical ImmunologyDepartment of Laboratory MedicineKarolinska InstituteHuddingeSweden
- Division of Biomedical EngineeringDepartment of Materials Science and EngineeringUppsala UniversityUppsalaAustralia
| |
Collapse
|
11
|
Kan L, Zheng Z, Fu W, Ma Y, Wang W, Qian H, Xu L. Recent progress on engineered micro/nanomaterials mediated modulation of gut microbiota for treating inflammatory bowel disease. J Control Release 2024; 370:43-65. [PMID: 38608876 DOI: 10.1016/j.jconrel.2024.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
Inflammatory bowel disease (IBD) is a type of chronic recurrent inflammation disease that mainly includes Crohn's disease and ulcerative colitis. Currently, the treatments for IBD remain highly challenging, with clinical treatment drugs showing limited efficacy and adverse side effects. Thus, developing drug candidates with comprehensive therapeutic effects, high efficiency, and low toxicity is urgently needed. Recently, micro/nanomaterials have attracted considerable interest because of their bioavailability, multitarget and efficient effects on IBD. In addition, gut modulation plays a substantial role in restoring intestinal homeostasis. Therefore, efficient microbiota-based strategies modulating gut microenvironment have great potential in remarkably treating IBD. With the development of micro- and nanomaterials for the treatment of IBD and more in-depth studies of their therapeutic mechanisms, it has been found that these treatments also have a tendency to positively regulate the intestinal flora, resulting in an increase in the beneficial flora and a decrease in the level of pathogenic bacteria, thus regulating the composition of the intestinal flora to a normal state. In this review, we first present the interactions among the immune system, intestinal barrier, and gut microbiome. In addition, recent advances in administration routes and methods that positively arouse the regulation of intestinal flora for IBD using probiotics, prebiotics, and redox-active micro/nanomaterials have been reviewed. Finally, the key challenges and critical perspectives of gut microbiota-based micro/nanomaterial treatment are also discussed.
Collapse
Affiliation(s)
- Lingling Kan
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China
| | - Ziwen Zheng
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China
| | - Wanyue Fu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China.
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China.
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, PR China.
| |
Collapse
|
12
|
Bai C, Liu J, Zhang X, Li Y, Qin Q, Song H, Yuan C, Huang Z. Research status and challenges of plant-derived exosome-like nanoparticles. Biomed Pharmacother 2024; 174:116543. [PMID: 38608523 DOI: 10.1016/j.biopha.2024.116543] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
In recent years, there has been an increasing number of related studies on exosomes. Most studies have focused on exosomes derived from mammals, confirming the important role that exosomes play in cell communication. Plants, as a natural ingredient, plant-derived exosomes have been confirmed to have similar structures and functions to mammalian-derived exosomes. Plant-derived exosome-like nanoparticles (PELNs) are lipid bilayer membrane nanovesicles containing bioactive constituents such as miRNA, mRNA, protein, and lipids obtained from plant cells, that can participate in intercellular communication and mediate transboundary communication, have high bioavailability and low immunogenicity, are relatively safe, and have been shown to play an important role in maintaining cell homeostasis and preventing, and treating a variety of diseases. In this review, we describe the biogenesis, isolation and purification methods, structural composition, stability, safety, function of PELNs and challenges. The functions of PELNs in anti-inflammatory, antioxidant, antitumor and drug delivery are mainly described, and the status of research on exosome nanoparticles of Chinese herbal medicines is outlined. Overall, we summarized the importance of PELNs and the latest research results in this field and provided a theoretical basis for the future research and clinical application of PELNs.
Collapse
Affiliation(s)
- Chunmei Bai
- The Fifth Clinical Medical College of Shanxi Medical University, Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, 030001, China
| | - Jianrong Liu
- The Fifth Clinical Medical College of Shanxi Medical University, Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, 030001, China; Department of reproductive medicine of Shanxi Provincial People's Hospital, Shuangtaxi Street, Taiyuan City, 030012, China.
| | - Xumin Zhang
- The Fifth Clinical Medical College of Shanxi Medical University, Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, 030001, China
| | - Yang Li
- The Fifth Clinical Medical College of Shanxi Medical University, Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, 030001, China
| | - Qin Qin
- The Fifth Clinical Medical College of Shanxi Medical University, Xinjian South Road, Yingze District, Taiyuan City, Shanxi Province, 030001, China; Department of reproductive medicine of Shanxi Provincial People's Hospital, Shuangtaxi Street, Taiyuan City, 030012, China
| | - Haixia Song
- Department of reproductive medicine of Shanxi Provincial People's Hospital, Shuangtaxi Street, Taiyuan City, 030012, China
| | - Caixia Yuan
- Department of reproductive medicine of Shanxi Provincial People's Hospital, Shuangtaxi Street, Taiyuan City, 030012, China
| | - Ziwei Huang
- Department of reproductive medicine of Shanxi Provincial People's Hospital, Shuangtaxi Street, Taiyuan City, 030012, China
| |
Collapse
|
13
|
Choi W, Cho JH, Park SH, Kim DS, Lee HP, Kim D, Kim HS, Kim JH, Cho JY. Ginseng root-derived exosome-like nanoparticles protect skin from UV irradiation and oxidative stress by suppressing activator protein-1 signaling and limiting the generation of reactive oxygen species. J Ginseng Res 2024; 48:211-219. [PMID: 38465216 PMCID: PMC10920011 DOI: 10.1016/j.jgr.2024.01.001] [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: 04/04/2023] [Revised: 12/05/2023] [Accepted: 01/10/2024] [Indexed: 03/12/2024] Open
Abstract
Background Recently, plant-derived exosome-like nanoparticles (PDENs) have been isolated, and active research was focusing on understanding their properties and functions. In this study, the characteristics and molecular properties of ginseng root-derived exosome-like nanoparticles (GrDENs) were examined in terms of skin protection. Methods HPLC-MS protocols were used to analyze the ginsenoside contents in GrDENs. To investigate the beneficial effect of GrDENs on skin, HaCaT cells were pre-treated with GrDENs (0-2 × 109 particles/mL), and followed by UVB irradiation or H2O2 exposure. In addition, the antioxidant activity of GrDENs was measured using a fluorescence microscope or flow cytometry. Finally, molecular mechanisms were examined with immunoblotting analysis. Results GrDENs contained detectable levels of ginsenosides (Re, Rg1, Rb1, Rf, Rg2 (S), Gyp17, Rd, C-Mc1, C-O, and F2). In UVB-irradiated HaCaT cells, GrDENs protected cells from death and reduced ROS production. GrDENs downregulated the mRNA expression of proapoptotic genes, including BAX, caspase-1, -3, -6, -7, and -8 and the ratio of cleaved caspase-8, -9, and -3 in a dose-dependent manner. In addition, GrDENs reduced the mRNA levels of aging-related genes (MMP2 and 3), proinflammatory genes (COX-2 and IL-6), and cellular senescence biomarker p21, possibly by suppressing activator protein-1 signaling. Conclusions This study demonstrates the protective effects of GrDENs against skin damage caused by UV and oxidative stress, providing new insights into beneficial uses of ginseng. In particular, our results suggest GrDENs as a potential active ingredient in cosmeceuticals to promote skin health.
Collapse
Affiliation(s)
- Wooram Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeong Hun Cho
- Research and Innovation Center, AMOREPACIFIC, Yongin, Republic of Korea
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dong Seon Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hwa Pyoung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Donghyun Kim
- Research and Innovation Center, AMOREPACIFIC, Yongin, Republic of Korea
| | - Hyun Soo Kim
- Research and Innovation Center, AMOREPACIFIC, Yongin, Republic of Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
14
|
Cao M, Diao N, Cai X, Chen X, Xiao Y, Guo C, Chen D, Zhang X. Plant exosome nanovesicles (PENs): green delivery platforms. MATERIALS HORIZONS 2023; 10:3879-3894. [PMID: 37671650 DOI: 10.1039/d3mh01030a] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Natural plants have been attracting increasing attention in biomedical research due to their numerous benefits. Plant exosome-derived vesicles, some of the plant's components, are small nanoscale vesicles secreted by plant cells. These vesicles are rich in bioactive substances and play significant roles in intercellular communication, information transfer, and maintaining homeostasis in organisms. They also hold promise for treating diseases, and their vesicular structures make them suitable carriers for drug delivery, with large-scale production feasible. Therefore, this paper aims to provide an overview of nanovesicles from different plant sources and their extraction methods. We also outline the biological activities of nanovesicles, including their anti-inflammatory, anti-viral, and anti-tumor properties, and systematically introduce their applications in drug delivery. These applications include transdermal delivery, targeted drug delivery, gene delivery, and their potential use in the modern food industry. This review provides new ideas and methods for future research on plant exosomes, including their empowerment by artificial intelligence and gene editing, as well as their potential application in the biomedicine, food, and agriculture industries.
Collapse
Affiliation(s)
- Min Cao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai 264005, P. R. China.
| | - Ningning Diao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai 264005, P. R. China.
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xing Chen
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| | - Yi Xiao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| | - Chunjing Guo
- College of Marine Life Science, Ocean University of China, 5# Yushan 10 Road, Qingdao 266003, P. R. China.
| | - Daquan Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai 264005, P. R. China.
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
15
|
Leng Y, Yang L, Zhu H, Li D, Pan S, Yuan F. Stability of Blueberry Extracellular Vesicles and Their Gene Regulation Effects in Intestinal Caco-2 Cells. Biomolecules 2023; 13:1412. [PMID: 37759813 PMCID: PMC10526224 DOI: 10.3390/biom13091412] [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: 09/05/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Plant extracellular vesicles (P-EVs) are considered promising functional food ingredients due to their various health benefits. In this study, blueberry extracellular vesicles (B-EVs) were collected and purified by size exclusion chromatography (SEC). The chemical compounds in B-EV extracts were analyzed by LC-MS/MS. In addition, the stability of B-EVs was evaluated during short- and long-term storage, heating, and in vitro digestion. The results showed that the B-EVs had a desirable particle size (88.2 ± 7.7 nm). Protein and total RNA concentrations were 582 ± 11.2 μg/mL and 15.4 μg/mL, respectively. The optimal storage temperatures for B-EVs were 4 °C and -80 °C for short- and long-term storage, respectively. Fluorescent labeling and qRT-PCR tests showed that B-EVs could be specifically internalized by Caco-2 cells, whereas virtually no cytotoxic or growth-inhibitory effects were observed. B-EVs down-regulated the expression levels of IL-1β and IL-8 and up-regulated the expression levels of NF-κβ and TLR5 in Caco-2 cells. Overall, the results proved that the intact structure of B-EVs could be preserved during food storage and processing conditions. B-EVs had the ability to reach the human intestine through oral delivery. Moreover, they could be absorbed by intestinal cells and affect human intestinal function.
Collapse
Affiliation(s)
- Yangfan Leng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.L.); (L.Y.); (H.Z.); (S.P.)
| | - Liubin Yang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.L.); (L.Y.); (H.Z.); (S.P.)
| | - Hangxin Zhu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.L.); (L.Y.); (H.Z.); (S.P.)
| | - Dongqin Li
- National Key Laboratory of Crop Genetic Improvement, National Center of Plant Gene Research (Wuhan), Huazhong Agricultural University, Wuhan 430070, China;
| | - Siyi Pan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.L.); (L.Y.); (H.Z.); (S.P.)
- Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control, Huazhong Agricultural University, Wuhan 430070, China
| | - Fang Yuan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.L.); (L.Y.); (H.Z.); (S.P.)
- Hubei Key Laboratory of Fruit & Vegetable Processing & Quality Control, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|