1
|
Ruan J, Xia Y, Ma Y, Xu X, Luo S, Yi J, Wu B, Chen R, Wang H, Yu H, Yang Q, Wu W, Sun D, Zhong J. Milk-derived exosomes as functional nanocarriers in wound healing: Mechanisms, applications, and future directions. Mater Today Bio 2025; 32:101715. [PMID: 40242483 PMCID: PMC12003018 DOI: 10.1016/j.mtbio.2025.101715] [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: 02/06/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Wound healing presents a significant challenge in healthcare, imposing substantial physiological and economic burdens. While traditional treatments and stem cell therapies have shown benefits, milk-derived exosomes (MDEs) offer distinct advantages as a cell-free therapeutic approach. MDEs, isolated from mammalian milk, are characterized by their biocompatibility, ease of acquisition, and high yield, making them a promising tool for enhancing wound repair. This review provides a comprehensive analysis of the composition, sources, and extraction methods of MDEs, with a focus on their therapeutic role in both acute and diabetic chronic wounds. MDEs facilitate wound healing through the delivery of bioactive molecules, modulating key processes such as inflammation, angiogenesis, and collagen synthesis. Their ability to regulate complex wound-healing pathways underscores their potential for widespread clinical application. This review highlights the importance of MDEs in advancing wound management and proposes strategies to optimize their use in regenerative medicine.
Collapse
Affiliation(s)
- Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yuping Xia
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| | - Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Xiyao Xu
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| | - Shihao Luo
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong Special Administrative Region of China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, 999077, Hong Kong Special Administrative Region of China
| | - Honggang Yu
- Hand and Foot Surgery, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China
- Jin Feng Laboratory, Chongqing, 401329, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Junbo Zhong
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| |
Collapse
|
2
|
Wang H, Wang Y, Ling M, Wang S, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Comparison of the Immunomodulatory Roles of Porcine Milk-Derived Extracellular Vesicles from Different Stages of Lactation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40327365 DOI: 10.1021/acs.jafc.4c11983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Milk-derived extracellular vesicles (MEV) have received increasing attention due to their physiological benefits for newborn growth and development. However, their physiological characteristics and immunomodulatory capacities during lactation remain unclear. In this study, we isolated MEV from porcine milk at day 1, week 1, week 2, and week 3 after parturition and compared their differences by common EV characterization, showing the higher concentration and smaller mean size of MEV from colostrum. Porcine MEV could be internalized by RAW264.7 macrophages and functionally deliver immune-related microRNAs (miRNAs). Furthermore, MEV promoted macrophage polarization toward the M2-like phenotype and exerted anti-inflammatory effects in vitro. Mice models with sepsis also displayed that MEV suppressed the secretion of pro-inflammatory cytokines in serum and drove an anti-inflammatory M2-like phenotype of the spleen, ultimately weakening immune responses. Especially, MEV from colostrum exhibited the most robust effects on immunoregulation. Overall, this study provides valuable information for MEV secretion patterns and their differences in immunomodulatory bioactivities.
Collapse
Affiliation(s)
- Hailong Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuxuan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Mingwang Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shumeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
3
|
Tong L, Wang Q, Zhang Y, Lai F, Xu J, Yin W, Zhang S, Wei G, Yin J, Yi H, Storm G, Wang Z, Huang R, Xu T, Wang JW. Myocardial delivery of miR30d with peptide-functionalized milk-derived extracellular vesicles for targeted treatment of hypertrophic heart failure. Biomaterials 2025; 316:122976. [PMID: 39637583 DOI: 10.1016/j.biomaterials.2024.122976] [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: 08/02/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
miR30d has been shown to reverse cardiac hypertrophy. However, effective delivery of miR30d to the heart is challenging. Here, we engineered milk-derived extracellular vesicles (mEVs) by surface functionalization with an ischemic myocardium-targeting peptide (IMTP) and encapsulated miR30d to develop a formulation, the miR30d-mEVsIMTP, enabling targeted delivery of miR30d to the injured heart. In vitro, the miR30d-mEVsIMTP can be effectively internalized by hypoxia-induced H9C2 cells via the endo-lysosomal pathway. In the isoproterenol (ISO)-induced cardiac hypertrophy mice, more miR30d-mEVsIMTP accumulated in cardiac tissue than miR30d-mEVs following intravenous administration. As a result, miR30d-mEVsIMTP alleviated cardiac hypertrophy and rescued cardiac function in three murine models of hypertrophic heart failure. Mechanistically, we identified GRK5 as an unprecedented target of miR30d in cardiac hypertrophy. Taken together, our findings demonstrate that mEVs conjugated with IMTP effectively deliver miR30d to the pathological heart and thereby ameliorating cardiac hypertrophy and dysfunction via targeting GRK5-mediated signaling pathways.
Collapse
Affiliation(s)
- Lingjun Tong
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qiyue Wang
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yameng Zhang
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Fengling Lai
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jiarun Xu
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wenchao Yin
- Department of Cardiology, Shandong First Medical Affiliated Shandong Provincial Hospital, Jinan, 250021, China
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore
| | - Guoyue Wei
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jie Yin
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Department of Cardiology, Shandong First Medical Affiliated Qianfoshan Hospital, Jinan 250013, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore
| | - Zhaoyang Wang
- Department of Cardiology, Shandong First Medical Affiliated Shandong Provincial Hospital, Jinan, 250021, China.
| | - Rong Huang
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Tao Xu
- Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250013, China; Guangzhou Laboratory, Guangzhou, 510005, China.
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117609, Singapore; Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS), Singapore, 117599, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
4
|
Farm YR, Chuah BH, Law JX, Leong XF, Razali M, Ng SL. Therapeutic Potential of Extracellular Vesicles in Oral Inflammation. Int J Mol Sci 2025; 26:3031. [PMID: 40243684 PMCID: PMC11988662 DOI: 10.3390/ijms26073031] [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: 02/21/2025] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
The therapeutic potential of extracellular vesicles (EVs) in reducing oral inflammation is thoroughly examined in this review, with an emphasis on gingivitis, periodontitis, and oral mucositis. It explains the complex relationship between microbial dysbiosis and host immune responses in the aetiology of oral inflammation. Pathophysiological mechanisms of periodontitis are examined, emphasising the roles played by periodontal pathogens and inflammatory mediators in the disease's chronic course and systemic effects. Preclinical research is providing new evidence that EVs originating from various cellular sources control immune cell dynamics towards a pro-healing phenotype, promote tissue regeneration, and have immunomodulatory qualities. EV-based therapies appear to be a promising new therapeutic technique with potential benefits over traditional methods for the treatment of oral inflammatory illnesses by specifically altering inflammatory signalling pathways. This review highlights the potential of EVs to improve patient outcomes in oral health and emphasises the need for additional clinical research to clarify the therapeutic efficacy and underlying mechanisms of EVs in periodontal therapy.
Collapse
Affiliation(s)
- Yan Rou Farm
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Bing Huan Chuah
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Xin Fang Leong
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Masfueh Razali
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| |
Collapse
|
5
|
Rahman MM, Inoshima Y. Prospects of bovine milk small extracellular vesicles in veterinary medicine. Res Vet Sci 2025; 184:105524. [PMID: 39765198 DOI: 10.1016/j.rvsc.2024.105524] [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: 11/27/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 02/01/2025]
Abstract
Extracellular vesicles (EV), including exosomes or small EV (sEV) derived from biological fluids, such as milk, have garnered increasing interest in veterinary medicine because of their role in the pathophysiology and understanding of the disease status of the host. Bovine milk serves as a rich source of sEV, containing diverse cargoes of nucleic acids, proteins, and lipids, which play a critical role in intercellular communication and regulation of host status. Although it is more difficult to isolate and purify sEV from bovine milk than from human breast milk, challenges persist in enabling the enrichment and analysis of sEV populations, facilitating the elucidation of their functional roles and prognostic potential in cattle diseases. Moreover, owing to their availability, ease of collection, noninvasive nature, and low cost, bovine milk sEV could be an excellent resource for research in veterinary medicine. Furthermore, the development of sEV-based prognosis is promising for improving veterinary medicine through the early detection of diseases and personalized therapeutic strategies. In this review, we provide a comprehensive overview of bovine milk sEV related to disease monitoring, host physiology, and immune regulation, and highlight their potential applications in advancing veterinary medicine. The prognostic and therapeutic potential of bovine milk sEV could be unlocked by combining knowledge from many fields, creating new opportunities for the development of precise, early prognostic, and focused therapeutic interventions for diseases in veterinary medicine.
Collapse
Affiliation(s)
- Md Matiur Rahman
- Laboratory of Food and Environmental Hygiene, Joint Department of Veterinary Medicine, Gifu University, Gifu, Japan; Department of Medicine, Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Yasuo Inoshima
- Laboratory of Food and Environmental Hygiene, Joint Department of Veterinary Medicine, Gifu University, Gifu, Japan; Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan; Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan.
| |
Collapse
|
6
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [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: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Budayr OM, Miller BC, Nguyen J. Harnessing extracellular vesicle-mediated crosstalk between T cells and cancer cells for therapeutic applications. J Control Release 2025; 378:266-280. [PMID: 39657892 PMCID: PMC11830559 DOI: 10.1016/j.jconrel.2024.12.011] [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: 07/17/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Small extracellular vesicles (EVs) are a diverse group of lipid-based particles that are ≤200 nm in diameter and contain an aqueous core. EVs have been shown to mediate intercellular communications between a wide array of immune cells; the downstream effects are diverse and have potential implications for the development of novel immunotherapeutic treatments. Despite a high volume of studies addressing the role EVs play in the immune system, our understanding of the crosstalk between T cells and cancer cells remains limited. Here, we discuss how EVs derived from cancer cells modulate T cell functions and conversely, how T cell derived EVs are crucial in modulating adaptive immune functions. In the context of cancer, tumor derived EVs (TD-EVs) halt T cell-mediated immunity by interfering with effector functions and enhancing regulatory T cell (Treg) functions. In contrast, EVs derived from effector T cells can serve to stimulate anticancer immunity, curbing metastasis and tumor growth. These findings highlight important aspects of how EVs can both mediate the therapeutic effects of T cells as well as impair T cell-mediated immunity. This calls for a deeper understanding of EV-mediated effects in order to advance them as next-generation therapeutics and nanocarriers.
Collapse
Affiliation(s)
- Omar M Budayr
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian C Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
8
|
Qu S, Yang S, Xu Q, Zhang M, Gao F, Wu Y, Li L. A Milk Extracellular Vesicle-Based Nanoplatform Enhances Combination Therapy Against Multidrug-Resistant Bacterial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406496. [PMID: 39721033 PMCID: PMC11831456 DOI: 10.1002/advs.202406496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/09/2024] [Indexed: 12/28/2024]
Abstract
The increasing occurrence of infections caused by multidrug-resistant (MDR) bacteria drives the need for new antibacterial drugs. Due to the current lack of antibiotic discovery and development, new strategies to fight MDR bacteria are urgently needed. Efforts to develop new antibiotic adjuvants to increase the effectiveness of existing antibiotics and design delivery systems are essential to address this issue. Here, a bioinspired delivery system equipped with combination therapy and paracellular transport is shown to enhance the efficacy against bacterial infections by improving oral delivery. A screening platform is established using an in vitro-induced high polymyxin-resistant strain to acquire plumbagin, which enhances the efficacy of polymyxin. Functionalized milk extracellular vesicles (FMEVs) coloaded with polymyxin and plumbagin cleared 99% of the bacteria within 4 h. Mechanistic studies revealed that the drug combination damaged the membrane, disrupted energy metabolism, and accelerated bacterial death. Finally, FMEVs are efficiently transported transcellularly through the citric acid-mediated reversible opening of the tight junctions and showed high efficacy against an MDR Escherichia coli-associated peritonitis-sepsis model in mice. These findings provide a potential therapeutic strategy to improve the efficacy of combination therapy by enhancing oral delivery using a biomimetic delivery platform.
Collapse
Affiliation(s)
- Shaoqi Qu
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
| | - Shuo Yang
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
| | - Qingjun Xu
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
| | - Mengying Zhang
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
| | - Feng Gao
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
| | - Yongning Wu
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
- Research Unit of Food SafetyChinese Academy of Medical Sciences (No. 2019RU014)NHC Key Laboratory of Food Safety Risk AssessmentChina National Center for Food Safety Risk Assessment (CFSA)Beijing100022China
| | - Lin Li
- Animal‐Derived Food Safety Innovation TeamCollege of Veterinary MedicineAnhui Agricultural UniversityHefei230036China
| |
Collapse
|
9
|
Marsh SR, Amin MR, Toldo S, Beard C, Dogan AB, Mezzaroma E, Andres E, Stout RF, Bannon MS, Payne LB, Abbate A, Sassi Y, Letteri RA, Gourdie RG. Orally Delivered Milk-Derived Nanovesicles Loaded with Connexin 43 Peptides for Targeted Cardiac Ischemia-Reperfusion Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.01.630994. [PMID: 40093162 PMCID: PMC11908194 DOI: 10.1101/2025.01.01.630994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Extracellular vesicles have emerged as promising nanocarriers for targeted drug delivery, but their therapeutic potential is limited by challenges related to administration route, loading, targeted delivery and production at scale. Here, we report an innovative approach for targeted delivery of therapeutic peptides to injured tissues using milk-derived small extracellular vesicles (mEVs) as an abundant, safe, orally administrable nanoplatform. We demonstrate that a sub-population of mEVs naturally contain Connexin 43 (Cx43) and its Carboxyl-Terminal (CT) polypeptides, which have been shown to play crucial roles in wound healing and tissue repair. Leveraging this intrinsic property, we developed an esterification method to efficiently and uniformly load mEVs with enhanced levels of an exogenous Cx43 CT peptide (αCT11 - RPRPDDLEI), as assessed by flow cytometry-based vesicle quantification and mass spectrometry. These engineered mEVs exhibited remarkable injury targeting capabilities, with > 30-fold increases in uptake by injured cells compared to non-wounded cells in vitro and preferential accumulation in wounded tissues in vivo. Notably, αCT11-loaded mEVs orally administered after myocardial infarction reduced infarct size by >60% and preserved heart function in a mouse model of ischemia-reperfusion injury. This study represents a significant advance in nanomedicine, demonstrating the utilization of naturally occurring milk-derived extracellular vesicles as an oral delivery system for therapeutic peptides, achieving unprecedented targeting efficiency and efficacy in the treatment of myocardial ischemia-reperfusion injury.
Collapse
|
10
|
Li Y, Wu SQ, Nan F, Deng W, Li K, Jarhen N, Zhou Y, Ma Q, Qu Y, Chen C, Ren Y, Yin XB. Single-Atom Iridium Nanozyme-Based Persistent Luminescence Nanoparticles for Multimodal Imaging-Guided Combination Tumor Therapy. Adv Healthc Mater 2024; 13:e2402544. [PMID: 39344246 DOI: 10.1002/adhm.202402544] [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] [Revised: 08/23/2024] [Indexed: 10/01/2024]
Abstract
Persistent luminescence nanoparticles (PLNPs) can achieve autofluorescence-free afterglow imaging, while near-infrared (NIR) emission realizes deep tissue imaging. Nanozymes integrate the merits of nanomaterials and enzyme-mimicking activities with simple preparation. Here PLNPs are prepared of Zn1.2Ga1.6Ge0.2O4:Cr0.0075 with NIR emission at 700 nm. The PLNPs are then incubated with IrCl3 solution, and the nanoparticles are collected and annealed at 750 °C to obtain iridium@PLNPs. Iridium is observed on the PLNPs at the atomic level as a single-atom nanozyme with peroxidase-like catalytic activity, photothermal conversion, and computed tomography (CT) contrast capability. After coating with exosome membrane (EM), the Ir@PLNPs@EM composite exhibits long-lasting NIR luminescence, peroxidase-like catalytic activity, photothermal conversion, and CT contrast capability, with the targeting capability and biocompatibility from EM. Thus, NIR afterglow/photothermal/CT trimodal imaging-guided photothermal-chemodynamic combination therapy is realized as validated with the in vitro and in vivo inhibition of tumor growth, while toxicity and side effects are avoided as drug-free treatment. This work offers a promising avenue for advanced single-atom nanozyme@PLNPs to promote the development of nanozymes and PLNPs for clinical applications.
Collapse
Affiliation(s)
- Yang Li
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shu-Qi Wu
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Fang Nan
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Wei Deng
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Kaixuan Li
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Nur Jarhen
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yitong Zhou
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Qianli Ma
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yuanyuan Qu
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Chaoxiang Chen
- Department of Biological Engineering, College of Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Yujing Ren
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing, 401135, China
| | - Xue-Bo Yin
- Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| |
Collapse
|
11
|
Joshi M, Sharma S, Onteru SK, Singh D. Comprehensive proteomic analysis of buffalo milk extracellular vesicles. Int J Biol Macromol 2024; 282:136735. [PMID: 39433188 DOI: 10.1016/j.ijbiomac.2024.136735] [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: 03/26/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
Extracellular vesicles are secretory vesicles involved in cell-to-cell communication via their encapsulated cargo of proteins, lipids, and nucleic acids. Bovine milk provides a rich source of extracellular vesicles (mEVs) that have been studied as therapeutics and drug delivery systems. Therefore, insight into the mEV cargo, such as its proteome, may help in understanding the molecular mechanism underlying the potential health benefits attributed to the mEVs. Hence, mEVs were isolated from healthy buffalo milk after screening the milk somatic cell count. The total proteins of mEVs were analyzed using LC-MS, and 331 proteins were found commonly present among three buffalo milk samples. These proteins were primarily derived from extracellular regions and lysosomes. The major biological roles associated with the proteins were immune response, metabolism, and cell cycle regulation. The molecular functions of the proteins were transporter activity, catalytic activity, and GTPase activity. Further, comparative analysis with the previously available bovine mEVs proteome data showed 114 proteins to be newly identified in the buffalo mEVs. The biological pathways associated with these proteins may play a major role in muscle development. These findings shed a light on the potential health benefits of buffalo mEVs as therapeutics as well as drug delivery vehicles.
Collapse
Affiliation(s)
- Mansi Joshi
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Sanjay Sharma
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India.
| |
Collapse
|
12
|
Saleem A, Saleem Bhat S, A. Omonijo F, A Ganai N, M. Ibeagha-Awemu E, Mudasir Ahmad S. Immunotherapy in mastitis: state of knowledge, research gaps and way forward. Vet Q 2024; 44:1-23. [PMID: 38973225 PMCID: PMC11232650 DOI: 10.1080/01652176.2024.2363626] [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: 11/02/2023] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
Mastitis is an inflammatory condition that affects dairy cow's mammary glands. Traditional treatment approaches with antibiotics are increasingly leading to challenging scenarios such as antimicrobial resistance. In order to mitigate the unwanted side effects of antibiotics, alternative strategies such as those that harness the host immune system response, also known as immunotherapy, have been implemented. Immunotherapy approaches to treat bovine mastitis aims to enhance the cow's immune response against pathogens by promoting pathogen clearance, and facilitating tissue repair. Various studies have demonstrated the potential of immunotherapy for reducing the incidence, duration and severity of mastitis. Nevertheless, majority of reported therapies are lacking in specificity hampering their broad application to treat mastitis. Meanwhile, advancements in mastitis immunotherapy hold great promise for the dairy industry, with potential to provide effective and sustainable alternatives to traditional antibiotic-based approaches. This review synthesizes immunotherapy strategies, their current understanding and potential future perspectives. The future perspectives should focus on the development of precision immunotherapies tailored to address individual pathogens/group of pathogens, development of combination therapies to address antimicrobial resistance, and the integration of nano- and omics technologies. By addressing research gaps, the field of mastitis immunotherapy can make significant strides in the control, treatment and prevention of mastitis, ultimately benefiting both animal and human health/welfare, and environment health.
Collapse
Affiliation(s)
- Afnan Saleem
- Division of Animal Biotechnology, SKUAST-K, Srinagar, India
| | | | - Faith A. Omonijo
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Canada
| | | | - Eveline M. Ibeagha-Awemu
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Canada
| | | |
Collapse
|
13
|
Barathan M, Ng SL, Lokanathan Y, Ng MH, Law JX. Milk-Derived Extracellular Vesicles: A Novel Perspective on Comparative Therapeutics and Targeted Nanocarrier Application. Vaccines (Basel) 2024; 12:1282. [PMID: 39591185 PMCID: PMC11599128 DOI: 10.3390/vaccines12111282] [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: 09/19/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Milk-derived extracellular vesicles (mEVs) are emerging as promising therapeutic candidates due to their unique properties and versatile functions. These vesicles play a crucial role in immunomodulation by influencing macrophage differentiation and cytokine production, potentially aiding in the treatment of conditions such as bone loss, fibrosis, and cancer. mEVs also have the capacity to modulate gut microbiota composition, which may alleviate the symptoms of inflammatory bowel diseases and promote intestinal barrier integrity. Their potential as drug delivery vehicles is significant, enhancing the stability, solubility, and bioavailability of anticancer agents while supporting wound healing and reducing inflammation. Additionally, bovine mEVs exhibit anti-aging properties and protect skin cells from UV damage. As vaccine platforms, mEVs offer advantages including biocompatibility, antigen protection, and the ability to elicit robust immune responses through targeted delivery to specific immune cells. Despite these promising applications, challenges persist, including their complex roles in cancer, effective antigen loading, regulatory hurdles, and the need for standardized production methods. Achieving high targeting specificity and understanding the long-term effects of mEV-based therapies are essential for clinical translation. Ongoing research aims to optimize mEV production methods, enhance targeting capabilities, and conduct rigorous preclinical and clinical studies. By addressing these challenges, mEVs hold the potential to revolutionize vaccine development and targeted drug delivery, ultimately improving therapeutic outcomes across various medical fields.
Collapse
Affiliation(s)
- Muttiah Barathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
14
|
Jiang Q, Wang L, Tian J, Zhang W, Cui H, Gui H, Zang Z, Li B, Si X. Food-derived extracellular vesicles: natural nanocarriers for active phytoconstituents in new functional food. Crit Rev Food Sci Nutr 2024; 64:11701-11721. [PMID: 37548408 DOI: 10.1080/10408398.2023.2242947] [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: 08/08/2023]
Abstract
Extracellular vesicles (EVs) are naturally occurring non-replicating particles released from cells, known for their health-promoting effects and potential as carriers for drug delivery. Extensive research has been conducted on delivery systems based on culture-cell-derived EVs. Nevertheless, they have several limitations including low production yield, high expenses, unsuitability for oral administration, and safety concerns in applications. Conversely, food-derived EVs (FDEVs) offer unique advantages that cannot be easily substituted. This review provides a comprehensive analysis of the biogenesis pathways, composition, and health benefits of FDEVs, as well as the techniques required for constructing oral delivery systems. Furthermore, it explores the advantages and challenges associated with FDEVs as oral nanocarriers, and discusses the current research advancements in delivering active phytoconstituents. FDEVs, functioning as a nanocarrier platform for the oral delivery of active molecules, present numerous benefits such as convenient administration, high biocompatibility, low toxicity, and inherent targeting. Nevertheless, numerous unresolved issues persist in the isolation, characterization, drug loading, and application of FDEVs. Technical innovation and standardization of quality control are the key points to promote the development of FDEVs. The review aimed to provide frontier ideas and basic quality control guidelines for developing new functional food based on FDEVs oral drug delivery system.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Weijia Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Hailong Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Zhihuan Zang
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| |
Collapse
|
15
|
Wang H, Wang Y, Wang S, Ling M, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Assessment of isolation strategies to remove caseins for high-quality milk-derived extracellular vesicles. J Dairy Sci 2024; 107:8934-8946. [PMID: 39033914 DOI: 10.3168/jds.2024-25162] [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: 05/13/2024] [Accepted: 06/27/2024] [Indexed: 07/23/2024]
Abstract
Increasing studies have highlighted the significance of milk-derived extracellular vesicles (MEV) in mother-newborn integration, as well as their application as novel drug delivery systems and diagnostic biomarkers. However, conventional ultracentrifugation (UC) often results in the co-precipitation of casein micelles in MEV pellets. In this study, we compared methods with different principles to screen the optimal pretreatment in caseins removal, and found that isoelectric precipitation by hydrochloric acid (HA) could most effectively remove caseins in porcine milk. We further characterized MEV populations isolated by UC and HA/UC from diverse aspects, including particle methodology via nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM), RNA and protein contents, and purity analysis. Importantly, the proliferative and anti-inflammatory effects of MEV were evaluated in vitro, showing the superiority of MEV via HA/UC in functionality compared with UC. Our results suggest that HA pretreatment before ultracentrifugation could effectively remove caseins and other protein complexes, making MEV with higher purity and more significant effects in vitro. This study provides valuable insights for the advancement of MEV isolation techniques across different species and accurate function analysis of MEV.
Collapse
Affiliation(s)
- Hailong Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuxuan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shumeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Mingwang Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
16
|
Lu L, Han C, Wang M, Du H, Chen N, Gao M, Wang N, Qi D, Bai W, Yin J, Dong F, Li T, Ge X. Assessment of bovine milk exosome preparation and lyophilized powder stability. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70009. [PMID: 39554868 PMCID: PMC11565256 DOI: 10.1002/jex2.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/18/2024] [Accepted: 08/30/2024] [Indexed: 11/19/2024]
Abstract
Exosomes are cell-derived nanovesicles that play a crucial role in intercellular communication, presenting promising potential as biomarkers and therapeutic agents. Bovine milk exosomes (MK-Exo) show production scalability and cost-effectiveness, offering distinct advantages over cell-derived exosomes. However, exosome storage and transportation are challenging owing to their unstable nature, necessitating preservation at ultralow temperatures. Research findings suggest that freeze-drying could provide a viable solution; however, different sources of exosomes may require specific protocols. In this study, we aimed to successfully isolate high-purity MK-Exo and develop a specialized freeze-drying and lyophilization method for improved long-term preservation of MK-Exo. Specifically, the stability of the lyophilized MK-Exo was evaluated using storage stability tests. Notably, lyophilized MK-Exo remained stable for at least 3 months under high temperature of 50°C and for at least 24 months under low temperatures of 2°C-8°C, preserving their physicochemical properties and biological activity. Conclusively, these findings provide a potential solution for ambient-temperature transportation of MK-Exo, facilitating their industrial-scale production.
Collapse
Affiliation(s)
- Lu Lu
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Chunle Han
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Miao Wang
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Huanqing Du
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Ning Chen
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Mengya Gao
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Na Wang
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Dongli Qi
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Wei Bai
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | - Jianxin Yin
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| | | | - Tianshi Li
- Pekin University Shenzhen HospitalShenzhenChina
| | - Xiaohu Ge
- Tingo Exosomes Technology Co. Ltd.TianjinChina
| |
Collapse
|
17
|
Ou H, Csuth TI, Czompoly T, Kvell K. Dairy: Friend or Foe? Bovine Milk-Derived Extracellular Vesicles and Autoimmune Diseases. Int J Mol Sci 2024; 25:11499. [PMID: 39519052 PMCID: PMC11546213 DOI: 10.3390/ijms252111499] [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: 09/23/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Due to the availability, scalability, and low immunogenicity, bovine milk-derived extracellular vesicles (MEVs) are increasingly considered to be a promising carrier of nanomedicines for future therapy. However, considering that extracellular vesicles (EVs) are of biological origin, different sources of EVs, including the host origin and the specific cells that produce the EVs, may have different effects on the structure and function of EVs. Additionally, MEVs play an important role in immune regulation, due to their evolutionary conserved cargo, such as cytokines and miRNAs. Their potential effects on different organs, as well as their accumulation in the human body, should not be overlooked. In this review, we have summarized current impacts and research progress brought about by utilizing MEVs as nano-drug carriers. Nevertheless, we also aim to explore the possible connections between the molecules involved in cellular immunity, cytokines and miRNAs of MEVs produced under different health conditions, and autoimmune diseases.
Collapse
Affiliation(s)
- Hairui Ou
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 7624 Pecs, Hungary; (H.O.); (T.I.C.); (K.K.)
| | - Tamas Imre Csuth
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 7624 Pecs, Hungary; (H.O.); (T.I.C.); (K.K.)
- Soft Flow Ltd., 7634 Pecs, Hungary
| | | | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 7624 Pecs, Hungary; (H.O.); (T.I.C.); (K.K.)
| |
Collapse
|
18
|
Xia B, Hu R, Chen J, Shan S, Xu F, Zhang G, Zhou Z, Fan Y, Hu Z, Liang XJ. Oral Administration Properties Evaluation of Three Milk-Derived Extracellular Vesicles Based on Ultracentrifugation Extraction Methods. Adv Healthc Mater 2024; 13:e2401370. [PMID: 38767497 DOI: 10.1002/adhm.202401370] [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: 04/15/2024] [Revised: 05/16/2024] [Indexed: 05/22/2024]
Abstract
Milk-derived extracellular vesicles (M-EVs) are low-cost, can be prepared in large quantities, and can cross the gastrointestinal barrier for oral administration. However, the composition of milk is complex, and M-EVs obtained by different extraction methods may affect their oral delivery. Based on this, a new method for extracting M-EVs based on cryogenic freezing treatment (Cryo-M-EVs) is proposed and compared with the previously reported acetic acid treatment (Acid-M-EVs) method and the conventional ultracentrifugation method (Ulltr-M-EVs). The new method simplifies the pretreatment step and achieves 25-fold and twofold higher yields than Acid-M-EVs and Ulltr-M-EVs. And it is interesting to note that Cryo-M-EVs and Acid-M-EVs have higher cellular uptake efficiency, and Cryo-M-EVs present the best transepithelial transport effect. After oral administration of the three M-EVs extracted by three methods in mice, Cryo-M-EVs effectively successfully cross the gastrointestinal barrier and achieve hepatic accumulation, whereas Acid-M-EVs and Ultr-M-EVs mostly reside in the intestine. The M-EVs obtained by the three extraction methods show a favorable safety profile at the cellular as well as animal level. Therefore, when M-EVs obtained by different extraction methods are used for oral drug delivery, their accumulation properties at different sites can be utilized to better deal with different diseases.
Collapse
Affiliation(s)
- Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Runjing Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junge Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing, 100083, China
| | - Shaobo Shan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, P. R. China
| | - Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Gang Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ziran Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing, 100083, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
19
|
Dai W, Zhou J, Chen T. Unraveling the extracellular vesicle network: insights into ovarian cancer metastasis and chemoresistance. Mol Cancer 2024; 23:201. [PMID: 39285475 PMCID: PMC11404010 DOI: 10.1186/s12943-024-02103-x] [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: 07/23/2024] [Accepted: 08/30/2024] [Indexed: 09/20/2024] Open
Abstract
Ovarian cancer (OC) is one of the most prevalent and lethal gynecological malignancies, with high mortality primarily due to its aggressive nature, frequent metastasis, and resistance to standard therapies. Recent research has highlighted the critical role of extracellular vesicles (EVs) in these processes. EVs, secreted by living organisms and carrying versatile and bioactive cargoes, play a vital role in intercellular communication. Functionally, the transfer of cargoes orchestrates multiple processes that actively affect not only the primary tumor but also local and distant pre-metastatic niche. Furthermore, their unique biological properties position EVs as novel therapeutic targets and promising drug delivery systems, with potential profound implications for cancer patients.This review summarizes recent progress in EV biology, delving into the intricate mechanisms by which EVs contribute to OC metastasis and drug resistance. It also explores the latest advances and therapeutic potential of EVs in the clinical context of OC. Despite the progress made, EV research in OC remains in its nascent stages. Consequently, this review presents existing research limitations and suggests avenues for future investigation. Altogether, the review aims to elucidate the critical roles of EVs in OC and spotlight their promising potential in this field.
Collapse
Affiliation(s)
- Wei Dai
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
- Zhejiang Provincial Clinical Research Center for CANCER, Hangzhou, Zhejiang, 310009, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Ting Chen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Zhejiang Provincial Clinical Research Center for CANCER, Hangzhou, Zhejiang, 310009, China.
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
20
|
Huang Q, Jiang Y, Cao Y, Ding Y, Cai J, Yang T, Zhou X, Wu Q, Li D, Liu Q, Li F. Bone-targeting engineered milk-derived extracellular vesicles for MRI-assisted therapy of osteoporosis. Regen Biomater 2024; 11:rbae112. [PMID: 39323741 PMCID: PMC11422186 DOI: 10.1093/rb/rbae112] [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: 06/18/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
The imbalance between osteoblasts and osteoclasts is the cause of osteoporosis. Milk-derived extracellular vesicles (mEVs), excellent drug delivery nanocarriers, can promote bone formation and inhibit bone resorption. In this study, we conjugated bone-targeting peptide (AspSerSer, DSS)6 to mEVs by click chemistry and then loaded with SRT2104, a SIRT1 (silent mating-type information regulation 2 homolog 1) agonist that was proofed to help reduce bone loss. The engineered (DSS)6-mEV-SRT2104 had the intrinsic anti-osteoporosis function of mEVs and SRT2104 to reverse the imbalance in bone homeostasis by simultaneously regulating osteogenesis and osteoclastogenesis. Furthermore, we labelled mEVs with MnB nanoparticles that can be used for the in vivo magnetic resonance imaging (MRI) visualization. The obtained nanocomposites significantly prevented bone loss in osteoporosis mice and increased bone mineral density, exhibiting superior bone accumulation under MRI. We believe the proposed (DSS)6-mEV-SRT2104/MnB provides a novel paradigm for osteoporosis treatment and monitoring.
Collapse
Affiliation(s)
- Qing Huang
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Jiang
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yang Cao
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yunchuan Ding
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinghui Cai
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Tingqian Yang
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Xin Zhou
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Qiang Wu
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Danyang Li
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Qingyu Liu
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Fangping Li
- Department of Endocrinology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
21
|
Chen C, Pan X, Sun M, Wang J, Su X, Zhang T, Chen Y, Wu D, Li J, Wu S, Yan X. Phospholipid-Anchored Ligand Conjugation on Extracellular Vesicles for Enhanced Cancer Targeting. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310712. [PMID: 38733222 DOI: 10.1002/smll.202310712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/27/2024] [Indexed: 05/13/2024]
Abstract
Extracellular vesicles (EVs) are recognized as potential candidates for next-generation drug delivery systems. However, the inherent cancer-targeting efficiency is unsatisfactory, necessitating surface modification to attach cell-binding ligands. By utilizing phospholipase D from Streptomyces in combination with maleimide-containing primary alcohol, the authors successfully anchored ligands onto milk-derived EVs (mEVs), overcoming the issues of ligand leakage or functional alteration seen in traditional methods. Quantitative nano-flow cytometry demonstrated that over 90% of mEVs are effectively modified with hundreds to thousands of ligands. The resulting mEV formulations exhibited remarkable long-term stability in conjugation proportion, ligand number, size distribution, and particle concentration, even after months of storage. It is further shown that conjugating transferrin onto mEVs significantly enhanced cellular uptake and induced pronounced cytotoxic effects when loaded with paclitaxel. Overall, this study presents a highly efficient, stable, cost-effective, and scalable ligand conjugation approach, offering a promising strategy for targeted drug delivery of EVs.
Collapse
Affiliation(s)
- Chaoxiang Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Xueping Pan
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Mengdi Sun
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Jialin Wang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Xueqi Su
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Tianyu Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Yulei Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Daren Wu
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Jian Li
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Shuqi Wu
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
22
|
Kim HK, Choi Y, Kim KH, Byun Y, Kim TH, Kim JH, An SH, Bae D, Choi MK, Lee M, Kang G, Chung J, Kim S, Kwon K. Scalable production of siRNA-encapsulated extracellular vesicles for the inhibition of KRAS-mutant cancer using acoustic shock waves. J Extracell Vesicles 2024; 13:e12508. [PMID: 39323378 PMCID: PMC11424982 DOI: 10.1002/jev2.12508] [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: 11/18/2023] [Revised: 07/16/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as a potential delivery vehicle for nucleic-acid-based therapeutics, but challenges related to their large-scale production and cargo-loading efficiency have limited their therapeutic potential. To address these issues, we developed a novel "shock wave extracellular vesicles engineering technology" (SWEET) as a non-genetic, scalable manufacturing strategy that uses shock waves (SWs) to encapsulate siRNAs in EVs. Here, we describe the use of the SWEET platform to load large quantities of KRASG12C-targeting siRNA into small bovine-milk-derived EVs (sBMEVs), with high efficiency. The siRNA-loaded sBMEVs effectively silenced oncogenic KRASG12C expression in cancer cells; they inhibited tumour growth when administered intravenously in a non-small cell lung cancer xenograft mouse model. Our study demonstrates the potential for the SWEET platform to serve as a novel method that allows large-scale production of cargo-loaded EVs for use in a wide range of therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - DaeHo Bae
- Exollence Co., Ltd.SeoulRepublic of Korea
| | | | | | - Gwansuk Kang
- Division of Gastroenterology and Hepatology, School of MedicineStanford UniversityStanfordCaliforniaUSA
| | | | | | - Kihwan Kwon
- Exollence Co., Ltd.SeoulRepublic of Korea
- Department of Internal Medicine, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| |
Collapse
|
23
|
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
|
24
|
Adamo G, Santonicola P, Picciotto S, Gargano P, Nicosia A, Longo V, Aloi N, Romancino DP, Paterna A, Rao E, Raccosta S, Noto R, Salamone M, Deidda I, Costa S, Di Sano C, Zampi G, Morsbach S, Landfester K, Colombo P, Wei M, Bergese P, Touzet N, Manno M, Di Schiavi E, Bongiovanni A. Extracellular vesicles from the microalga Tetraselmis chuii are biocompatible and exhibit unique bone tropism along with antioxidant and anti-inflammatory properties. Commun Biol 2024; 7:941. [PMID: 39097626 PMCID: PMC11297973 DOI: 10.1038/s42003-024-06612-9] [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: 10/03/2023] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-enclosed bio-nanoparticles secreted by cells and naturally evolved to transport various bioactive molecules between cells and even organisms. These cellular objects are considered one of the most promising bio-nanovehicles for the delivery of native and exogenous molecular cargo. However, many challenges with state-of-the-art EV-based candidates as drug carriers still exist, including issues with scalability, batch-to-batch reproducibility, and cost-sustainability of the final therapeutic formulation. Microalgal extracellular vesicles, which we named nanoalgosomes, are naturally released by various microalgal species. Here, we evaluate the innate biological properties of nanoalgosomes derived from cultures of the marine microalgae Tetraselmis chuii, using an optimized manufacturing protocol. Our investigation of nanoalgosome biocompatibility in preclinical models includes toxicological analyses, using the invertebrate model organism Caenorhabditis elegans, hematological and immunological evaluations ex vivo and in mice. We evaluate nanoalgosome cellular uptake mechanisms in C. elegans at cellular and subcellular levels, and study their biodistribution in mice with accurate space-time resolution. Further examination highlights the antioxidant and anti-inflammatory bioactivities of nanoalgosomes. This holistic approach to nanoalgosome functional characterization demonstrates that they are biocompatible and innate bioactive effectors with unique bone tropism. These findings suggest that nanoalgosomes have significant potential for future therapeutic applications.
Collapse
Affiliation(s)
- Giorgia Adamo
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Pamela Santonicola
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Sabrina Picciotto
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Paola Gargano
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Aldo Nicosia
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Valeria Longo
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Noemi Aloi
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Daniele P Romancino
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Angela Paterna
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Estella Rao
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Samuele Raccosta
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Rosina Noto
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Monica Salamone
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Irene Deidda
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Salvatore Costa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Caterina Di Sano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Palermo, Italy
| | - Giuseppina Zampi
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research (MPIP), Mainz, Germany
| | | | - Paolo Colombo
- Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
| | - Mingxing Wei
- Cellvax SAS, Villejuif Bio Park, 1 Mail du Professeur Georges Mathé, Villejuif, France
| | - Paolo Bergese
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Center for Colloid and Surface Science (CSGI), Florence, Italy
| | - Nicolas Touzet
- Department of Environmental Science, School of Science, Centre for Environmental Research, Innovation and Sustainability, CERIS, Atlantic Technological University Sligo, Sligo, Ireland
| | - Mauro Manno
- Cell-Tech HUB at Institute of Biophysics, National Research Council of Italy (CNR), Palermo, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Antonella Bongiovanni
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo, Italy.
| |
Collapse
|
25
|
Wang J, Yin B, Lian J, Wang X. Extracellular Vesicles as Drug Delivery System for Cancer Therapy. Pharmaceutics 2024; 16:1029. [PMID: 39204374 PMCID: PMC11359799 DOI: 10.3390/pharmaceutics16081029] [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: 06/17/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
In recent decades, the pursuit of drug delivery systems has led to the development of numerous synthetic options aimed at enhancing drug efficacy while minimizing side effects. However, the practical application of these systems is often hindered by challenges such as inefficiency, cytotoxicity, and immunogenicity. Extracellular vesicles, natural carriers for drugs, emerge as promising alternatives with distinct advantages over synthetic carriers. Notably, EVs exhibit biocompatibility, low immunogenicity, and inherent tissue-targeting capabilities, thus opening new avenues for drug delivery strategies. This review provides an overview of EVs, including their biogenesis and absorption mechanisms. Additionally, we explore the current research efforts focusing on harnessing their potential as drug carriers, encompassing aspects such as purification techniques, drug loading, and bioengineering for targeted delivery. Finally, we discuss the existing challenges and future prospects of EVs as therapeutic agents in clinical settings. This comprehensive analysis aims to shed light on the potential of EVs as versatile and effective tools for drug delivery, particularly in the realm of cancer therapy.
Collapse
Affiliation(s)
- Jin Wang
- School of Life Sciences, Liaoning University, Shenyang 110036, China; (J.W.); (J.L.)
| | - Bohang Yin
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Jiabing Lian
- School of Life Sciences, Liaoning University, Shenyang 110036, China; (J.W.); (J.L.)
| | - Xia Wang
- Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenyang 110122, China
| |
Collapse
|
26
|
Xu Q, Yang S, Zhang K, Liu Y, Li L, Qu S. Enhanced antibacterial activity of bovine milk exosome-based drug formulation against bacterial pathogens. Food Chem 2024; 447:139034. [PMID: 38493686 DOI: 10.1016/j.foodchem.2024.139034] [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: 11/29/2023] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Milk is not only a source of nutrients, but also contains exosomes (Exo) that can serve as a vehicle for drug delivery. Here, we obtained bovine milk Exo using three efficient methods, demonstrating high quality for commercial production. The optimized Exo displayed a size of 105.2 nm and an entrapment efficiency of 88.4 %. The Exo has been functionalized with a combination therapy comprising isobavachalcone (IS) and polymyxin B (PB), referred to as IP-Exo. The antibacterial efficacy of IP-Exo was significantly enhanced, enabling the elimination of 99 % of multidrug-resistant (MDR) bacterial pathogens in 4 h. Furthermore, scanning electron microscopy images demonstrated that the drug combination led to the complete dismantling of the bacterial structure. IP-Exo showed nearly 100 % microbial inhibition in fresh orange juice and accelerated wound healing in mouse models. Collectively, IP-Exo provides excellent potential for application within the food industry and animal husbandry as a defense against bacterial pathogens.
Collapse
Affiliation(s)
- Qingjun Xu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shuo Yang
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Kai Zhang
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
27
|
Bahadorani M, Nasiri M, Dellinger K, Aravamudhan S, Zadegan R. Engineering Exosomes for Therapeutic Applications: Decoding Biogenesis, Content Modification, and Cargo Loading Strategies. Int J Nanomedicine 2024; 19:7137-7164. [PMID: 39050874 PMCID: PMC11268655 DOI: 10.2147/ijn.s464249] [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: 03/04/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Exosomes emerge from endosomal invagination and range in size from 30 to 200 nm. Exosomes contain diverse proteins, lipids, and nucleic acids, which can indicate the state of various physiological and pathological processes. Studies have revealed the remarkable clinical potential of exosomes in diagnosing and prognosing multiple diseases, including cancer, cardiovascular disorders, and neurodegenerative conditions. Exosomes also have the potential to be engineered and deliver their cargo to a specific target. However, further advancements are imperative to optimize exosomes' diagnostic and therapeutic capabilities for practical implementation in clinical settings. This review highlights exosomes' diagnostic and therapeutic applications, emphasizing their engineering through simple incubation, biological, and click chemistry techniques. Additionally, the loading of therapeutic agents onto exosomes, utilizing passive and active strategies, and exploring hybrid and artificial exosomes are discussed.
Collapse
Affiliation(s)
- Mehrnoosh Bahadorani
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Mahboobeh Nasiri
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Shyam Aravamudhan
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Reza Zadegan
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| |
Collapse
|
28
|
Wu X, Shen J, Zhong Y, Zhao X, Zhou W, Gao P, Wang X, An W. Large-Scale Isolation of Milk Exosomes for Skincare. Pharmaceutics 2024; 16:930. [PMID: 39065627 PMCID: PMC11279399 DOI: 10.3390/pharmaceutics16070930] [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: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Exosomes are small membrane vesicles in a cell culture. They are secreted by most cells and originate from the endosomal pathway. A variety of proteins, lipids, and genetic materials have been shown to be carried by exosomes. Once taken up by neighboring or distant cells, the bioactive compounds in exosomes can regulate the condition of recipient cells. Typically, producing exosomes in large quantities requires cell culture, resulting in high production costs. However, exosomes are abundant in milk and can be isolated on a large scale at a low cost. In our study, we found that milk exosomes can promote the synthesis and reconstruction of stratum corneum lipids, enhance skin barrier function, and provide greater protection for the skin. Furthermore, milk exosomes have anti-inflammatory properties that can reduce skin irritation, redness, and other symptoms, giving immediate relief. They also exhibit antioxidant activity, which helps neutralize free radicals and slows down the skin aging process. Additionally, milk exosomes inhibit melanin production, aiding in skin whitening. Ongoing research has uncovered the benefits of milk exosomes for skin improvement and their application in cosmetics, skin healthcare, and other fields, and these applications are continuing to expand.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xudong Wang
- China National Biotech Group (CNBG), Sinopharm Group, National Vaccine & Serum Institute (NVSI), No. 38 Jing Hai Second Road, Beijing 101111, China; (X.W.); (J.S.); (Y.Z.); (X.Z.); (W.Z.); (P.G.)
| | - Wenlin An
- China National Biotech Group (CNBG), Sinopharm Group, National Vaccine & Serum Institute (NVSI), No. 38 Jing Hai Second Road, Beijing 101111, China; (X.W.); (J.S.); (Y.Z.); (X.Z.); (W.Z.); (P.G.)
| |
Collapse
|
29
|
Kumar J, Onteru SK, Singh D. Deciphering the Drug Delivery Potential of Milk Exosome Nanovesicles for Aminobenzylpenicillin Therapeutic Efficacy against Contagious Staphylococcus Aureus in Bovine Mastitis. Adv Biol (Weinh) 2024; 8:e2300519. [PMID: 38573624 DOI: 10.1002/adbi.202300519] [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: 09/29/2023] [Revised: 01/21/2024] [Indexed: 04/05/2024]
Abstract
The emergence of antimicrobial resistance and failure of antibiotic treatment are challenging tasks for managing bovine mastitis, which is mainly caused by the contagious Staphylococcus aureus (S. aureus).To overcome these difficulties, there is an urgent need for a novel drug system. In the present study, the aim is to develop next-generation therapeutics against S. aureus by harnessing the drug delivery potential of milk nanovesicles called milk exosomes (mENs). In the present work, a drug system is developed by encapsulating aminobenzylpenicillin (AMP) in mENs (mENs-AMP). Electron microscopy and zeta-sizer results indicate that the size of mENs-AMP ranged from 55.79 ± 2.8 to 85.53 ± 7.4 nm. The AMP loading efficiency in mENs is 88.61% with its sustained release. Fluorescence spectroscopy results indicated that mENs are biocompatible with mammary epithelial cells. In vitro studies show that the antibacterial activity and the minimum inhibitory concentrations of mENs-AMP are eleven times greater and four times lower than that of unencapsulated AMP, respectively. The mENs-AMP exhibit significantly higher therapeutic efficacy than AMP at the same dosage and treatment frequency. Validation of this approach is demonstrated in mastitis-affected animals through an observation in the reduction of somatic cell counts and bacterial loads in the milk of treated animals.
Collapse
Affiliation(s)
- Jitendra Kumar
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
30
|
Zhang S, Wang Q, Tan DEL, Sikka V, Ng CH, Xian Y, Li D, Muthiah M, Chew NWS, Storm G, Tong L, Wang J. Gut-liver axis: Potential mechanisms of action of food-derived extracellular vesicles. J Extracell Vesicles 2024; 13:e12466. [PMID: 38887165 PMCID: PMC11183959 DOI: 10.1002/jev2.12466] [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: 01/13/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Food-derived extracellular vesicles (FEVs) are nanoscale membrane vesicles obtained from dietary materials such as breast milk, plants and probiotics. Distinct from other EVs, FEVs can survive the harsh degrading conditions in the gastrointestinal tract and reach the intestines. This unique feature allows FEVs to be promising prebiotics in health and oral nanomedicine for gut disorders, such as inflammatory bowel disease. Interestingly, therapeutic effects of FEVs have recently also been observed in non-gastrointestinal diseases. However, the mechanisms remain unclear or even mysterious. It is speculated that orally administered FEVs could enter the bloodstream, reach remote organs, and thus exert therapeutic effects therein. However, emerging evidence suggests that the amount of FEVs reaching organs beyond the gastrointestinal tract is marginal and may be insufficient to account for the significant therapeutic effects achieved regarding diseases involving remote organs such as the liver. Thus, we herein propose that FEVs primarily act locally in the intestine by modulating intestinal microenvironments such as barrier integrity and microbiota, thereby eliciting therapeutic impact remotely on the liver in non-gastrointestinal diseases via the gut-liver axis. Likewise, drugs delivered to the gastrointestinal system through FEVs may act via the gut-liver axis. As the liver is the main metabolic hub, the intestinal microenvironment may be implicated in other metabolic diseases. In fact, many patients with non-alcoholic fatty liver disease, obesity, diabetes and cardiovascular disease suffer from a leaky gut and dysbiosis. In this review, we provide an overview of the recent progress in FEVs and discuss their biomedical applications as therapeutic agents and drug delivery systems, highlighting the pivotal role of the gut-liver axis in the mechanisms of action of FEVs for the treatment of gut disorders and metabolic diseases.
Collapse
Affiliation(s)
- Sitong Zhang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Qiyue Wang
- Jinan Central HospitalShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Daniel En Liang Tan
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Vritika Sikka
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cheng Han Ng
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingaporeSingapore
| | - Yan Xian
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dan Li
- Department of Food Science and Technology, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingaporeSingapore
- National University Centre for Organ TransplantationNational University Health SystemSingaporeSingapore
| | - Nicholas W. S. Chew
- Department of CardiologyNational University Heart CentreNational University Health SystemSingaporeSingapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Lingjun Tong
- Jinan Central HospitalShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Jiong‐Wei Wang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Cardiovascular Research Institute (CVRI)National University Heart Centre Singapore (NUHCS)SingaporeSingapore
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
31
|
Jiang Q, Liu Y, Si X, Wang L, Gui H, Tian J, Cui H, Jiang H, Dong W, Li B. Potential of Milk-Derived Extracellular Vesicles as Carriers for Oral Delivery of Active Phytoconstituents. Annu Rev Food Sci Technol 2024; 15:431-454. [PMID: 38359948 DOI: 10.1146/annurev-food-072023-034354] [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: 02/17/2024]
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication and have the potential to serve as in vivo carriers for delivering active molecules. The biocompatibility advantages of EVs over artificial nanocarriers create new frontiers for delivering modern active molecules. Milk is a favorable source of EVs because of its high bioavailability, low immunogenicity, and commercial producibility. In this review, we analyzed the advantages of milk-derived EVs in the oral delivery of active molecules, discussed their research progress in delivering active phytoconstituents, and summarized the necessary technologies and critical unit operations required for the development of an oral delivery system based on EVs. The review aims to provide innovative ideas and fundamental quality control guidelines for developing the next-generation oral drug delivery system based on milk-derived EVs.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Yubo Liu
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hailong Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Hongzhou Jiang
- Anhui Ziyue Biological Technology Co., Ltd., Wuhu, China
| | - Wenjiang Dong
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| |
Collapse
|
32
|
Leone I, Santoro J, Soricelli A, Febbraro A, Santoriello A, Carrese B. Triple-Negative Breast Cancer EVs Modulate Growth and Migration of Normal Epithelial Lung Cells. Int J Mol Sci 2024; 25:5864. [PMID: 38892050 PMCID: PMC11172765 DOI: 10.3390/ijms25115864] [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: 04/24/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancer is the most common cancer amongst women worldwide. Recently, owing to screening programs and new technologies, the survival rate has increased significantly. Breast cancer can potentially develop metastases, and, despite them, lung metastases generally occur within five years of breast cancer diagnosis. In this study, the objective was to analyze the effect of breast cancer-derived EVs on a lung epithelial cell line. BEAS-2B cells were treated with extracellular vesicles (EVs) derived from triple-negative breast cancer cells (TNBCs), e.g., MDA-MB-231 and HS578T, separated using differential ultracentrifugation. We observed an increased growth, migration, and invasiveness of normal epithelial lung cells over time in the presence of TNBC EVs compared to the control. Therefore, these data suggest that EVs released by tumor cells contain biological molecules capable of influencing the pro-tumorigenic activity of normal cells. Exploring the role of EVs in oncology research and their potential cargo may be novel biomarkers for early cancer detection and further diagnosis.
Collapse
Affiliation(s)
- Ilaria Leone
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| | - Andrea Soricelli
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| | - Antonio Febbraro
- Oncology Unit, Casa di Cura Cobellis, Vallo della Lucania, 84078 Vallo della Lucania, Italy;
| | - Antonio Santoriello
- Breast Unit, Casa di Cura Cobellis, Vallo della Lucania, 84078 Vallo della Lucania, Italy;
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via E. Gianturco, 80143 Naples, Italy; (I.L.); (A.S.); (B.C.)
| |
Collapse
|
33
|
Cetinkaya H, Kongsomros S, Nommsen-Rivers L, Morrow AL, Chutipongtanate S. Which casein micelle removal method is suitable for studies of human milk extracellular vesicles? A systematic comparison of four different treatments for casein depletion before extracellular vesicle isolation from human milk. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:221-232. [PMID: 39698537 PMCID: PMC11648521 DOI: 10.20517/evcna.2024.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/16/2024] [Accepted: 05/14/2024] [Indexed: 12/20/2024]
Abstract
Aim: This study aimed to systematically compare four casein micelle removal methods on the particle and protein characteristics of the isolated human milk EVs. Methods: The defatted milk was treated with 1% sodium citrate, 20 mM ethylenediaminetetraacetic acid (EDTA), 1% acetic acid, or 1% chymosin/calcium chloride for 30 min at 4 °C to remove casein micelles. EV isolation was performed using qEV size exclusion chromatography. Milk turbidity at the optical density 350 nm and dot immunoblot with casein antibody were applied to monitor the qEV fractions. Particle analyses were performed using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). The enrichment of human milk EV markers, i.e., tetraspanins, Alix, lactadherin, butyrophilin, and xanthine dehydrogenase, and casein depletion capabilities were evaluated by proteomics and immunoblotting. Results: Compared to the untreated condition, sodium citrate and EDTA decreased milk turbidity by disrupting casein micelles, while acetic acid and chymosin removed them by inducing precipitation/coagulation. All treatments shifted casein immunoreactivity in the qEV fractions from large micelles (the exclusion volume) to small molecular sizes (gel-infiltrated fractions). Acidification affected human milk EV morphology, while EDTA, acetic acid, and chymosin methods slightly altered EV particle numbers. Different casein micelle removal methods confer different degrees of human milk EV marker enrichment and casein depletion. The method performances could be ranked as follows: chymosin > EDTA > acetic acid > sodium citrate. Conclusion: Our findings suggest that chymosin and EDTA should be considered as the method of choice for casein micelle removal in future studies involving human milk EV isolation and characterization.
Collapse
Affiliation(s)
- Hatice Cetinkaya
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Authors contributed equally
| | - Supasek Kongsomros
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Authors contributed equally
| | | | - Ardythe L. Morrow
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Somchai Chutipongtanate
- MILCH and Novel Therapeutics Lab, Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
34
|
Turner NP. Food-derived extracellular vesicles in the human gastrointestinal tract: Opportunities for personalised nutrition and targeted therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e154. [PMID: 38939572 PMCID: PMC11080705 DOI: 10.1002/jex2.154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/25/2024] [Accepted: 04/20/2024] [Indexed: 06/29/2024]
Abstract
Food-derived extracellular vesicles (FDEVs) such as those found in mammalian milk and plants are of great interest for both their health benefits and ability to act as biological nanocarriers. While the extracellular vesicle (EV) field is expanding rapidly to perform characterisation studies on FDEVs from plants, yeasts and bacteria, species-specific differences in EV uptake and function in the human gastrointestinal (GI) tract are poorly understood. Moreover, the effects of food processing on the EV surfaceome and intraluminal content also raises questions surrounding biological viability once consumed. Here, I present a case for increasing community-wide focus on understanding the cellular uptake of FDEVs from different animal, plant, yeast, and bacterial species and how this may impact their function in the human, which will have implications for human health and therapeutic strategies alike.
Collapse
Affiliation(s)
- Natalie P. Turner
- Faculty of HealthQueensland University of TechnologyKelvin GroveQueenslandAustralia
| |
Collapse
|
35
|
Salehi M, Negahdari B, Mehryab F, Shekari F. Milk-Derived Extracellular Vesicles: Biomedical Applications, Current Challenges, and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8304-8331. [PMID: 38587896 DOI: 10.1021/acs.jafc.3c07899] [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: 04/09/2024]
Abstract
Extracellular vesicles (EVs) are nano to-micrometer-sized sacs that are released by almost all animal and plant cells and act as intercellular communicators by transferring their cargos between the source and target cells. As a safe and scalable alternative to conditioned medium-derived EVs, milk-derived EVs (miEVs) have recently gained a great deal of popularity. Numerous studies have shown that miEVs have intrinsic therapeutic actions that can treat diseases and enhance human health. Additionally, they can be used as natural drug carriers and novel classes of biomarkers. However, due to the complexity of the milk, the successful translation of miEVs from benchtop to bedside still faces several unfilled gaps, especially a lack of standardized protocols for the isolation of high-purity miEVs. In this work, by comprehensively reviewing the bovine miEVs studies, we provide an overview of current knowledge and research on miEVs while highlighting their challenges and enormous promise as a novel class of theranostics. It is hoped that this study will pave the way for clinical applications of miEVs by addressing their challenges and opportunities.
Collapse
Affiliation(s)
- Mahsa Salehi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
| | - Fatemeh Mehryab
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 14155-6153, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| |
Collapse
|
36
|
Wang K, Zhao X, Yang S, Qi X, Zang G, Li C, Li A, Chen B. Milk-derived exosome nanovesicles: recent progress and daunting hurdles. Crit Rev Food Sci Nutr 2024; 65:2388-2403. [PMID: 38595109 DOI: 10.1080/10408398.2024.2338831] [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: 04/11/2024]
Abstract
Raw milk is the foundation of quality and safety in the dairy industry, and improving milk source management is the fundamental guarantee. Milk-derived exosomes (MDEs) are nanoscale information transfer molecules secreted by mammary cells with unique content and high stability, which can be used not only as potential markers to analyze key traits of lactation, reproduction, nutrition and health of animals, but also help farm managers to take timely interventions to improve animal welfare, milk quality, and functional traits. Our review first outlines the latest advances in MDEs isolation and purification, compositional analysis and characterization tools. We then provide a comprehensive summary of recent applications of MDEs liquid biopsy in breed selection, disease prevention and control, and feeding management. Finally, we evaluate the impact of processing on the stability of MDEs to offer guidance for dairy production and storage. The limitations and challenges in the development and use of MDEs markers are also discussed. As a noninvasive marker with high sensitivity and specificity, the MDEs-mediated assay technology is expected to be a powerful tool for measuring cow health and raw milk quality, enabling dynamic and precise regulation of dairy cows and full traceability of raw milk.
Collapse
Affiliation(s)
- Kaili Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Xu Zhao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Sijia Yang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Xiaoxi Qi
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Guofang Zang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Chun Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Research Institute, Harbin, China
| | - Aili Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Research Institute, Harbin, China
| | - Bingcan Chen
- Department of Plant Sciences, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|
37
|
Dai C, Xu Q, Li L, Liu Y, Qu S. Milk Extracellular Vesicles: Natural Nanoparticles for Enhancing Oral Drug Delivery against Bacterial Infections. ACS Biomater Sci Eng 2024; 10:1988-2000. [PMID: 38529792 DOI: 10.1021/acsbiomaterials.3c01824] [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: 03/27/2024]
Abstract
Oral drug delivery is typically preferred as a therapeutic intervention due to the complexities and expenses associated with intravenous administration. However, some drugs are poorly absorbed orally, requiring intravenous administration to bypass the gastrointestinal tract and deliver the drug directly into the bloodstream. Thus, there is an urgent need to develop novel drug delivery platforms to overcome the challenges of oral drug delivery with low solubility, low permeability, oral degradation, and low bioavailability. Advances in extracellular vesicles (EVs) as natural carriers have provided emerging approaches to improve potential therapeutic applications. Milk not only contains traditional nutrients but is also rich in EVs. In this Review, we focus mainly on the purification of milk EVs (mEVs), their safety, and the advantages of mEV-based drug carriers in combatting intestinal infections. Additionally, we summarize several advantages of mEVs over conventional synthetic carriers, such as low immunogenicity, high biocompatibility, and the ability to transfer bioactive molecules between cells. Considering the unmet gaps of mEVs in clinical translation, it is essential to review the cargo loading into mEVs and future perspectives for their use as natural drug carriers for oral delivery. This overview of mEV-based drug carriers for oral delivery sheds light on alternative approaches to treat clinical infections associated with intestinal pathogens and the development of novel oral delivery systems.
Collapse
Affiliation(s)
- Cunchun Dai
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Qingjun Xu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
38
|
Kankaanpää S, Nurmi M, Lampimäki M, Leskinen H, Nieminen A, Samoylenko A, Vainio SJ, Mäkinen S, Ahonen L, Kangasluoma J, Petäjä T, Viitala S. Comparative analysis of the effects of different purification methods on the yield and purity of cow milk extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e149. [PMID: 38938848 PMCID: PMC11080921 DOI: 10.1002/jex2.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 02/21/2024] [Accepted: 03/29/2024] [Indexed: 06/29/2024]
Abstract
Isolation of extracellular vesicles (EV) has been developing rapidly in parallel with the interest in EVs. However, commonly utilized protocols may not suit more challenging sample matrixes and could potentially yield suboptimal results. Knowing and assessing the pitfalls of isolation procedure to be used, should be involved to some extent for EV analytics. EVs in cow milk are of great interest due to their abundancy and large-scale availability as well as their cross-species bioavailability and possible use as drug carriers. However, the characteristics of milk EVs overlap with those of other milk components. This makes it difficult to isolate and study EVs individually. There exists also a lack of consensus for isolation methods. In this study, we demonstrated the differences between various differential centrifugation-based approaches for isolation of large quantities of EVs from cow milk. Samples were further purified with gradient centrifugation and size exclusion chromatography (SEC) and differences were analyzed. Quality measurements were conducted on multiple independent platforms. Particle analysis, electron microscopy and RNA analysis were used, to comprehensively characterize the isolated samples and to identify the limitations and possible sources of contamination in the EV isolation protocols. Vesicle concentration to protein ratio and RNA to protein ratios were observed to increase as samples were purified, suggesting co-isolation with major milk proteins in direct differential centrifugation protocols. We demonstrated a novel size assessment of vesicles using a particle mobility analyzer that matched the sizing using electron microscopy in contrast to commonly utilized nanoparticle tracking analysis. Based on the standards of the International Society for Extracellular Vesicles and the quick checklist of EV-Track.org for EV isolation, we emphasize the need for complete characterization and validation of the isolation protocol with all EV-related work to ensure the accuracy of results and allow further analytics and experiments.
Collapse
Affiliation(s)
| | - Markus Nurmi
- Natural Resources Institute FinlandJokioinenFinland
| | - Markus Lampimäki
- Institute for Atmospheric and Earth System Research (INAR) / PhysicsUniversity of HelsinkiHelsinkiFinland
| | | | - Anni Nieminen
- Metabolomics Unit, Institute for Molecular Medicine FinlandUniversity of HelsinkiHelsinkiFinland
| | - Anatoliy Samoylenko
- University of Oulu, Kvantum Institute, Infotech Oulu, Faculty of Biochemistry and Molecular Medicine, Disease Networks Research UnitOulu UniversityOuluFinland
| | - Seppo J. Vainio
- University of Oulu, Kvantum Institute, Infotech Oulu, Faculty of Biochemistry and Molecular Medicine, Disease Networks Research UnitOulu UniversityOuluFinland
| | - Sari Mäkinen
- Natural Resources Institute FinlandJokioinenFinland
| | - Lauri Ahonen
- Institute for Atmospheric and Earth System Research (INAR) / PhysicsUniversity of HelsinkiHelsinkiFinland
| | - Juha Kangasluoma
- Institute for Atmospheric and Earth System Research (INAR) / PhysicsUniversity of HelsinkiHelsinkiFinland
| | - Tuukka Petäjä
- Institute for Atmospheric and Earth System Research (INAR) / PhysicsUniversity of HelsinkiHelsinkiFinland
| | | |
Collapse
|
39
|
Santoro J, Nuzzo S, Franzese M, Salvatore M, Grimaldi AM. Goat milk extracellular vesicles: Separation comparison of natural carriers for theragnostic application. Heliyon 2024; 10:e27621. [PMID: 38509910 PMCID: PMC10950560 DOI: 10.1016/j.heliyon.2024.e27621] [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: 09/26/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
Goat milk is a complex biological fluid, which in addition to having a high nutritional value, it is an interesting source of extracellular vesicles (EVs). Despite the countless potential applications that they offer in many biological fields, is not easy to compare the different proposed systems, and this is a major limitation for the real translatability of these natural nanoplatforms for theragnostic purposes. Thus, it is useful to further investigate reproducible methods to separate goat milk EVs. The choice of methods but also the preprocessing of milk has an immense impact on the separation, quality, and yield of EVs. Here, we tested four protocols to separate EVs from unpasteurised goat milk: two based on differential ultracentrifugation (DUC) and two on size-exclusion chromatography (SEC). Moreover, we assessed two different approaches of pre-treatment (acidification and precipitation) to facilitate milk protein discharge. To the best of our knowledge, a similar comparison of all performed protocols on raw goat milk has never been published before. Therefore, enriched EV samples were successfully obtained from goat milk using both DUC and SEC. Taken together, our results may be helpful to obtain natural carriers for future theragnostic applications in personalised medicine.
Collapse
Affiliation(s)
- Jessie Santoro
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143, Napoli, Italy
| | - Silvia Nuzzo
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143, Napoli, Italy
| | - Monica Franzese
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143, Napoli, Italy
| | - Marco Salvatore
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143, Napoli, Italy
| | | |
Collapse
|
40
|
Liu Y, Luo Z, Xie Y, Sun Y, Yuan F, Jiang L, Lu H, Hu J. Extracellular vesicles from UTX-knockout endothelial cells boost neural stem cell differentiation in spinal cord injury. Cell Commun Signal 2024; 22:155. [PMID: 38424563 PMCID: PMC10903014 DOI: 10.1186/s12964-023-01434-4] [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: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Vascular endothelial cells are pivotal in the pathophysiological progression following spinal cord injury (SCI). The UTX (Ubiquitously Transcribed Tetratripeptide Repeat on Chromosome X) serves as a significant regulator of endothelial cell phenotype. The manipulation of endogenous neural stem cells (NSCs) offers a compelling strategy for the amelioration of SCI. METHODS Two mouse models were used to investigate SCI: NSCs lineage-traced mice and mice with conditional UTX knockout (UTX KO) in endothelial cells. To study the effects of UTX KO on neural differentiation, we harvested extracellular vesicles (EVs) from both UTX KO spinal cord microvascular endothelial cells (SCMECs) and negative control SCMECs. These EVs were then employed to modulate the differentiation trajectory of endogenous NSCs in the SCI model. RESULTS In our NSCs lineage-traced mice model of SCI, a marked decrease in neurogenesis was observed post-injury. Notably, NSCs in UTX KO SCMECs mice showed enhanced neuronal differentiation compared to controls. RNA sequencing and western blot analyses revealed an upregulation of L1 cell adhesion molecule (L1CAM), a gene associated with neurogenesis, in UTX KO SCMECs and their secreted EVs. This aligns with the observed promotion of neurogenesis in UTX KO conditions. In vivo administration of L1CAM-rich EVs from UTX KO SCMECs (KO EVs) to the mice significantly enhanced neural differentiation. Similarly, in vitro exposure of NSCs to KO EVs resulted in increased activation of the Akt signaling pathway, further promoting neural differentiation. Conversely, inhibiting Akt phosphorylation or knocking down L1CAM negated the beneficial effects of KO EVs on NSC neuronal differentiation. CONCLUSIONS In conclusion, our findings substantiate that EVs derived from UTX KO SCMECs can act as facilitators of neural differentiation following SCI. This study not only elucidates a novel mechanism but also opens new horizons for therapeutic interventions in the treatment of SCI. Video Abstract.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zixiang Luo
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Xie
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Sun
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Feifei Yuan
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyuan Jiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Jianzhong Hu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
41
|
Furukawa S, Kawaguchi K, Chikama K, Yamada R, Kamatari YO, Lim LW, Koyama H, Inoshima Y, Ikemoto MJ, Yoshida S, Hirata Y, Furuta K, Takemori H. Simple methods for measuring milk exosomes using fluorescent compound GIF-2250/2276. Biochem Biophys Res Commun 2024; 696:149505. [PMID: 38219490 DOI: 10.1016/j.bbrc.2024.149505] [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: 11/30/2023] [Revised: 12/24/2023] [Accepted: 01/08/2024] [Indexed: 01/16/2024]
Abstract
Exosomes are small extracellular vesicles (EVs) found in culture supernatants, blood, and breast milk. The size of these nanocomplexes limits the methods of EV analyses. In this study, nitrobenzoxadiazole (NBD), a fluorophore, conjugated endosome-lysosome imager, GIF-2250 and its derivative, GIF-2276, were evaluated for exosome analyses. A correlation was established between GIF-2250 intensity and protein maker levels in bovine milk exosomes. We found that high-temperature sterilization milk may not contain intact exosomes. For precise analysis, we synthesized GIF-2276, which allows for the covalent attachment of NBD to the Lys residue of exosome proteins, and labeled milk exosomes were separated using a gel filtration system. GIF-2276 showed chromatographic peaks of milk exosomes containing >3 ng protein. The area (quantity) and retention time (size) of the exosome peaks were correlated to biological activity (NO synthesis suppression in RAW264.7 murine macrophages). Heat denaturation of purified milk-derived exosomes disrupted these indicators. Proteome analyses revealed GIF-2276-labeled immunomodulators, such as butyrophilin subfamily 1 member A1 and polymeric immunoglobulin receptor. The immunogenicity and quantity of these factors decreased by heat denaturation. When milk exosomes were purified from market-sourced milk we found that raw and low-temperature sterilization milk samples, contained exosomes (none in high-temperature sterilization milk). These results were also supported by transmission electron microscopy analyses. We also found that GIF-2276 could monitor exosome transportation into HEK293 cells. These results suggested that GIF-2250/2276 may be helpful to evaluate milk exosomes.
Collapse
Affiliation(s)
- Saho Furukawa
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kyoka Kawaguchi
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kotomi Chikama
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Ryohei Yamada
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yuji O Kamatari
- Life Science Research Center, Gifu University, Gifu, Gifu, 501-1193, Japan; The United Graduate School of Drug Discovery and Medical Information Sciences of Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Lee Wah Lim
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Hiroko Koyama
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan; The United Graduate School of Drug Discovery and Medical Information Sciences of Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yasuo Inoshima
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Mitsushi J Ikemoto
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan; Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sports Sciences, University of Tsukuba, Tsukuba, 305-8574, Japan
| | - Saishi Yoshida
- Seki Gyunyu Co. Ltd, 41, Kannonmae, Seki, Gifu, 501-3835, Japan
| | - Yoko Hirata
- Life Science Research Center, Gifu University, Gifu, Gifu, 501-1193, Japan
| | - Kyoji Furuta
- GIFU EXOSOME Co. Ltd, 1-11-9, Yabuta-minani, Gifu, 500-8384, Japan
| | - Hiroshi Takemori
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan; The United Graduate School of Drug Discovery and Medical Information Sciences of Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan; GIFU EXOSOME Co. Ltd, 1-11-9, Yabuta-minani, Gifu, 500-8384, Japan.
| |
Collapse
|
42
|
Kumar MA, Baba SK, Sadida HQ, Marzooqi SA, Jerobin J, Altemani FH, Algehainy N, Alanazi MA, Abou-Samra AB, Kumar R, Al-Shabeeb Akil AS, Macha MA, Mir R, Bhat AA. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther 2024; 9:27. [PMID: 38311623 PMCID: PMC10838959 DOI: 10.1038/s41392-024-01735-1] [Citation(s) in RCA: 166] [Impact Index Per Article: 166.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 02/06/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous structures secreted into the extracellular space. They exhibit diverse sizes, contents, and surface markers and are ubiquitously released from cells under normal and pathological conditions. Human serum is a rich source of these EVs, though their isolation from serum proteins and non-EV lipid particles poses challenges. These vesicles transport various cellular components such as proteins, mRNAs, miRNAs, DNA, and lipids across distances, influencing numerous physiological and pathological events, including those within the tumor microenvironment (TME). Their pivotal roles in cellular communication make EVs promising candidates for therapeutic agents, drug delivery systems, and disease biomarkers. Especially in cancer diagnostics, EV detection can pave the way for early identification and offers potential as diagnostic biomarkers. Moreover, various EV subtypes are emerging as targeted drug delivery tools, highlighting their potential clinical significance. The need for non-invasive biomarkers to monitor biological processes for diagnostic and therapeutic purposes remains unfulfilled. Tapping into the unique composition of EVs could unlock advanced diagnostic and therapeutic avenues in the future. In this review, we discuss in detail the roles of EVs across various conditions, including cancers (encompassing head and neck, lung, gastric, breast, and hepatocellular carcinoma), neurodegenerative disorders, diabetes, viral infections, autoimmune and renal diseases, emphasizing the potential advancements in molecular diagnostics and drug delivery.
Collapse
Affiliation(s)
- Mudasir A Kumar
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Sadaf K Baba
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Sara Al Marzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Faisal H Altemani
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad A Alanazi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
43
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 1069] [Impact Index Per Article: 1069.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
44
|
Huang M, Shao H, Zhang X, Yang F, Wang J, Tan S, Chen H, Li X. Comparison of cow's milk allergy models highlighted higher humoral and Th2 immune responses in BALB/c than C3H/HeNCrl mice. Food Chem Toxicol 2024; 184:114315. [PMID: 38081529 DOI: 10.1016/j.fct.2023.114315] [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: 09/10/2023] [Revised: 11/09/2023] [Accepted: 11/28/2023] [Indexed: 12/25/2023]
Abstract
Cow's milk allergy (CMA) is common in early childhood and the incidence is increasing. However, its mechanisms of action are still not fully understood due to the range of different clinical symptoms. So far, the development of different mouse models has been the best choice to study the molecular mechanisms triggering allergy. However, the selection of suitable strains for the establishment of animal models truly representative of associated human pathologies is still a challenge. Hence, we focused on both C3H/HeNCrl and BALB/c mice to characterize their susceptibility to CMA. After intraperitoneal sensitization, BALB/c and C3H/HeNCrl strains were challenged with β-lactoglobulin (BLG), and compared in allergic symptoms and active immune response, which assessed by specific antibody production and cytokine release. At first, both groups exhibited anaphylaxis, showed specific BLG-related IgE, Th2 response and seemed both suitable for the development of CMA models. However, a detailed analysis revealed that BALB/c had both stronger humoral and Th2 immune responses, producing more antibodies (IgE and IgG/IgG1/IgG2a), and releasing higher levels of Th2-associated cytokines (IL-4, IL-5, IL-13) compared to C3H/HeNCrl mice. Therefore, BALB/c strain would represent a preferential choice in the establishment of CMA models. This study highlights the subtle differences and major outcomes in the selection of mouse strains for the development of suitable food allergy models.
Collapse
Affiliation(s)
- Meijia Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science, Henan Institute of Science and Technology, Xinxiang, 453003, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Huming Shao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Fan Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Jingshu Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Shuijie Tan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; Sino-German Joint Research Institute, Nanchang University, Nanchang, 330047, Jiangxi, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, Jiangxi, PR China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, PR China; School of Food Science and Technology, Nanchang University, Nanchang, 330031, Jiangxi, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, 330047, Jiangxi, PR China.
| |
Collapse
|
45
|
Zhang C, Zhang H, Millán Cotto HA, Boyer TL, Warren MR, Wang CM, Luchan J, Dhal PK, Carrier RL, Bajpayee AG. Milk exosomes anchored with hydrophilic and zwitterionic motifs enhance mucus permeability for applications in oral gene delivery. Biomater Sci 2024; 12:634-649. [PMID: 38047368 PMCID: PMC10842862 DOI: 10.1039/d3bm01089a] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Exosomes have emerged as a promising tool for the delivery of drugs and genetic materials, owing to their biocompatibility and non-immunogenic nature. However, challenges persist in achieving successful oral delivery due to their susceptibility to degradation in the harsh gastrointestinal (GI) environment and impeded transport across the mucus-epithelium barrier. To overcome these challenges, we have developed high-purity bovine milk exosomes (mExo) as a scalable and efficient oral drug delivery system, which can be customized by incorporating hydrophilic and zwitterionic motifs on their surface. In our study, we observed significantly improved transport rates by 2.5-4.5-fold in native porcine intestinal mucus after the introduction of hydrophilic and zwitterionic surface modifications, as demonstrated by transwell setup and fluorescence recovery after photobleaching (FRAP) analysis. Remarkably, mExo functionalized by a block peptide (BP), consisting of cationic and anionic amino acids arranged in blocks at the two ends, demonstrated superior tolerability in the acidic gastric environment (with a protein recovery rate of 84.8 ± 7.7%) and exhibited a 2.5-fold increase in uptake by intestinal epithelial cells. Furthermore, both mExo and mExo-BP demonstrated successful intracellular delivery of functional siRNA, resulting in up to 65% suppression of the target green fluorescence protein (GFP) gene expression at a low dose of siRNA (5 pmol) without causing significant toxicity. These findings highlight the immense potential of modifying mExo with hydrophilic and zwitterionic motifs for effective oral delivery of siRNA therapies.
Collapse
Affiliation(s)
- Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | | | - Timothy L Boyer
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Matthew R Warren
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Joshua Luchan
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | | | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
46
|
Hirayu N, Takasu O. Exploring the Hemostatic Effects of Platelet Lysate-Derived Vesicles: Insights from Mouse Models. Int J Mol Sci 2024; 25:1188. [PMID: 38256259 PMCID: PMC10816445 DOI: 10.3390/ijms25021188] [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: 11/20/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Platelet transfusion has various challenges, and platelet-derived extracellular vesicles have been reported to have more significant procoagulant activity than platelets themselves. Furthermore, platelet products derived from platelet-rich plasma and platelet lysates (PLs) have gained attention for their physiological activity and potential role as drug delivery vehicles owing to the properties of their membranes. We aimed to investigate the characteristics of the fractions isolated through ultracentrifugation from mouse-washed PLs and assess the potential clinical applications of these fractions as a therapeutic approach for bleeding conditions. We prepared PLs from C57BL/6 mouse-washed platelets and isolated three different fractions (20K-vesicles, 100K-vesicles, and PLwo-vesicles) using ultracentrifugation. There was a notable difference in particle size distribution between 20K-vesicles and 100K-vesicles, particularly in terms of the most frequent diameter. The 20K-vesicles exhibited procoagulant activity with concentration dependence, whereas PLwo-vesicles exhibited anticoagulant activity. PLwo-vesicles did not exhibit thrombin generation capacity, and the addition of PLwo-vesicles to Microparticle Free Plasma extended the time to initiate thrombin generation by 20K-vesicles and decreased the peak thrombin value. In a tail-snip bleeding assay, pre-administration of 20K-vesicles significantly shortened bleeding time. PL-derived 20K-vesicles exhibited highly potent procoagulant activity, making them potential alternatives to platelet transfusion.
Collapse
Affiliation(s)
- Nobuhisa Hirayu
- Department of Emergency and Critical Care Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan;
| | | |
Collapse
|
47
|
Ferruggia G, Contino M, Zimbone M, Brundo MV. Effectiveness of a Novel Compound HAIR & SCALP COMPLEX on Hair Follicle Regeneration. COSMETICS 2024; 11:10. [DOI: 10.3390/cosmetics11010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Background: People lose between 50 and 100 hairs a day and generate new ones from stem cells in hair follicles, but in those suffering from baldness, the stem cells remain inactive and are unable to regenerate new hair. Although 9% of hair follicles remain in telogen at any time, a variety of factors, including growth factors and cytokines, promote the transition from telogen to anagen and the subsequent stimulation of hair growth. Methods: We compared in vitro, on cultures of human hair follicles, the effect on hair growth and regeneration of the dermal papilla of plant-derived nanovesicles, exosomes from cord blood stem cells and bovine colostrum, a mixture of growth factors and cytokines purified from bovine colostrum, called GF20, and a new compound called HAIR & SCALP COMPLEX obtained by adding exosomes isolated from colostrum to GF20. Results: The analyses demonstrated a significant increase in the growth of the bulb and the regeneration of the dermal papilla in the samples treated with HAIR & SCALP COMPLEX compared to the other elements tested. Conclusions: In this research, we propose a possible new treatment that could help significantly slow down hair loss and encourage new hair growth: HAIR & SCALP COMPLEX.
Collapse
Affiliation(s)
- Greta Ferruggia
- Department of Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Martina Contino
- Department of Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Massimo Zimbone
- Institute for Microelectronics and Microsystems, National Research Council of Italy (CNR-IMM), 95123 Catania, Italy
| | - Maria Violetta Brundo
- Department of Biological, Geological and Environmental Sciences, University of Catania, Via Androne 81, 95124 Catania, Italy
| |
Collapse
|
48
|
Mondal J, Pillarisetti S, Junnuthula V, Surwase SS, Hwang SR, Park IK, Lee YK. Extracellular vesicles and exosome-like nanovesicles as pioneering oral drug delivery systems. Front Bioeng Biotechnol 2024; 11:1307878. [PMID: 38260737 PMCID: PMC10800420 DOI: 10.3389/fbioe.2023.1307878] [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: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
As extracellular vesicle (EV)-based nanotechnology has developed rapidly, it has made unprecedented opportunities for nanomedicine possible. EVs and exosome-like nanovesicles (ELNVs) are natural nanocarriers with unique structural, compositional, and morphological characteristics that provide excellent physical, chemical, and biochemical properties. In this literature, we examine the characteristics of EVs, including how they are administered orally and their therapeutic activity. According to the current examples of EVs and ELNVs for oral delivery, milk and plant EVs can exert therapeutic effects through their protein, nucleic acid, and lipid components. Furthermore, several methods for loading drugs into exosomes and targeting exosomes have been employed to investigate their therapeutic capability. Moreover, we discuss EVs as potential drug carriers and the potential role of ELNVs for disease prevention and treatment or as potential drug carriers in the future. In conclusion, the issues associated with the development of EVs and ELNVs from sources such as milk and plants, as well as concerns with standardized applications of these EVs, are discussed.
Collapse
Affiliation(s)
- Jagannath Mondal
- Department of Green Bioengineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Shameer Pillarisetti
- Department of Biomedical Sciences and Biomedical Science Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | | | - Sachin S. Surwase
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and Biomedical Science Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yong-kyu Lee
- Department of Green Bioengineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
49
|
Colella AP, Prakash A, Miklavcic JJ. Homogenization and thermal processing reduce the concentration of extracellular vesicles in bovine milk. Food Sci Nutr 2024; 12:131-140. [PMID: 38268886 PMCID: PMC10804120 DOI: 10.1002/fsn3.3749] [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: 05/01/2023] [Revised: 07/19/2023] [Accepted: 09/20/2023] [Indexed: 01/26/2024] Open
Abstract
Extracellular vesicles (EVs) in bovine milk confer beneficial physiologic effects to consumers. Industrial processing treatments may affect the amount or bioactivity of EVs intrinsic to bovine milk. We investigated how the content and concentration of EVs were affected by homogenization and thermal processing of raw bovine milk. Raw milk was processed by homogenization, low-temperature (LT) heat, or pasteurization [high-temperature short-time (HTST) and ultra-high-temperature (UHT)] in a pilot processing facility. EVs were isolated from the raw and processed bovine milk using differential ultracentrifugation and quantified using a nanoparticle tracking analyzer. Bovine milk EVs were assessed for total miRNA and protein concentrations standardized to particle count using a fluorometric assay. There were 1.01 × 1010 (±3.30 × 109) EV particles per ml of bovine milk. All industrial processing treatments caused >60% decrease in EV concentration compared to the raw bovine milk. Homogenization and heat treatments independently and additively reduced the content of EVs in bovine milk. The averages of total miRNA/particle and total protein/particle concentrations were elevated threefold by low-temperature heat-processing treatment relative to HTST and UHT pasteurizations. The average diameter of EVs was reduced by 11%-16% by low temperature compared to raw milk (127 ± 13 nm). Homogenization and pasteurization indiscriminately reduce the EV concentration of bovine milk. Smaller EVs with higher protein content resist degradation when processing bovine milk at sub-pasteurization temperature. This new foundational knowledge may contribute to food product development on the preservation of EVs in processed dairy products, including bovine milk-based infant formulas that some newborns are dependent on for adequate growth and development.
Collapse
Affiliation(s)
- Anna P. Colella
- Schmid College of Science and TechnologyChapman UniversityOrangeCaliforniaUSA
| | - Anuradha Prakash
- Schmid College of Science and TechnologyChapman UniversityOrangeCaliforniaUSA
| | - John J. Miklavcic
- Schmid College of Science and TechnologyChapman UniversityOrangeCaliforniaUSA
- School of PharmacyChapman UniversityIrvineCaliforniaUSA
| |
Collapse
|
50
|
Stastna M. Advances in separation and identification of biologically important milk proteins and peptides. Electrophoresis 2024; 45:101-119. [PMID: 37289082 DOI: 10.1002/elps.202300084] [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: 04/26/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
Milk is a rich source of biologically important proteins and peptides. In addition, milk contains a variety of extracellular vesicles (EVs), including exosomes, that carry their own proteome cargo. EVs are essential for cell-cell communication and modulation of biological processes. They act as nature carriers of bioactive proteins/peptides in targeted delivery during various physiological and pathological conditions. Identification of the proteins and protein-derived peptides in milk and EVs and recognition of their biological activities and functions had a tremendous impact on food industry, medicine research, and clinical applications. Advanced separation methods, mass spectrometry (MS)-based proteomic approaches and innovative biostatistical procedures allowed for characterization of milk protein isoforms, genetic/splice variants, posttranslational modifications and their key roles, and contributed to novel discoveries. This review article discusses recently published developments in separation and identification of bioactive proteins/peptides from milk and milk EVs, including MS-based proteomic approaches.
Collapse
Affiliation(s)
- Miroslava Stastna
- Institute of Analytical Chemistry of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|