1
|
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
|
2
|
Wang J, Zhang Y, Wang S, Wang X, Jing Y, Su J. Bone aging and extracellular vesicles. Sci Bull (Beijing) 2024; 69:3978-3999. [PMID: 39455324 DOI: 10.1016/j.scib.2024.10.013] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Bone aging, a major global health concern, is the natural decline in bone mass and strength. Concurrently, extracellular vesicles (EVs), tiny membrane-bound particles produced by cells, have gained recognition for their roles in various physiological processes and age-related diseases. The interaction between EVs and bone aging is of growing interest, particularly their effects on bone metabolism, which become increasingly critical with advancing age. In this review, we explored the biology, types, and functions of EVs and emphasized their regulatory roles in bone aging. We examined the effects of EVs on bone metabolism and highlighted their potential as biomarkers for monitoring bone aging progression. Furthermore, we discussed the therapeutic applications of EVs, including targeted drug delivery and bone regeneration, and addressed the challenges associated with EV-based therapies, including the technical complexities and regulatory issues. We summarized the current research and clinical trials investigating the role of EVs in bone aging and suggested future research directions. These include the potential for personalized medicine using EVs and the integration of EV research with advanced technologies to enhance the management of age-related bone health. This analysis emphasized the transformative potential of EVs in understanding and managing bone aging, thereby marking a significant advancement in skeletal health research.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; School of Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
3
|
Di S, Huang Y, Qiao W, Zhang X, Wang Y, Zhang M, Fu J, Zhao J, Chen L. Advances in the isolation and characterization of milk-derived extracellular vesicles and their functions. Front Nutr 2024; 11:1512939. [PMID: 39742102 PMCID: PMC11688093 DOI: 10.3389/fnut.2024.1512939] [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: 10/17/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Milk-derived extracellular vesicles (EVs) have various functions, including immune regulation and promoting intestinal development. These EVs have substantial potential for application in infant formula and functional foods development. In addition, numerous studies have shown that milk-derived EVs carry proteins, lipids, and nucleic acids away from their parental cells, acting as messengers between cells. Moreover, structural integrity and biological viability are necessary prerequisites for the functional and omics studies of milk-derived EVs. Therefore, selecting appropriate methods for isolating and characterizing milk-derived EVs is essential for subsequent studies. Accordingly, this review summarizes the isolation and characterization methods for milk-derived EVs and their biological functions and roles. Furthermore, it discusses the comprehensive application of isolation methods, providing a reference for research on and development of milk-derived EVs.
Collapse
Affiliation(s)
- Shujuan Di
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Yibo Huang
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Weicang Qiao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Xiaomei Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Yaling Wang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Minghui Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Jieyu Fu
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Junying Zhao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lijun Chen
- Key Laboratory of Dairy Science, Ministry of Education, Food Science College, Northeast Agricultural University, Harbin, China
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| |
Collapse
|
4
|
Hu Y, Cui J, Sun J, Liu X, Gao S, Mei X, Wu C, Tian H. A novel biomimetic nanovesicle containing caffeic acid-coupled carbon quantum dots for the the treatment of Alzheimer's disease via nasal administration. J Nanobiotechnology 2024; 22:642. [PMID: 39425199 PMCID: PMC11490022 DOI: 10.1186/s12951-024-02912-8] [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: 07/23/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by progressive cognitive and physical impairment. Neuroinflammation is related to AD, and the misfolding and aggregation of amyloid protein in the brain creates an inflammatory microenvironment. Microglia are the predominant contributors to neuroinflammation, and abnormal activation of microglia induces the release of a large amount of inflammatory factors, promotes neuronal apoptosis, and leads to cognitive impairment. In this study, we used microglial membranes containing caffeic acid-coupled carbon quantum dots to prepare a novel biomimetic nanocapsule (CDs-CA-MGs) for the treatment of AD. The application of CDs-CA-MGs via nasal administration can bypass the blood‒brain barrier (BBB) and directly target the site of inflammation. After treatment with CDs-CA-MGs, AD mice showed reduced inflammation in the brain, decreased neuronal apoptosis, and significantly improved learning and memory abilities. In addition, CDs-CA-MGs affect inflammation-related JAK-STAT and Toll-like receptor signaling pathways in AD mice. CDs-CA-MGs significantly downregulated interleukins (IL-1β and IL-6) and tumor necrosis factor (TNF-α). This finding suggested that CDs-CA-MGs may improve cognitive impairment by modulating inflammatory responses. In conclusion, the use of CDs-CA-MGs provides a possible therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jingwen Cui
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Junpeng Sun
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xiaobang Liu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Shuang Gao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xifan Mei
- Liaoning Vocational College of Medicine, Shenyang, Liaoning, 110101, China.
| | - Chao Wu
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| | - He Tian
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
5
|
Yu T, Zhao IS, Pan H, Yang J, Wang H, Deng Y, Zhang Y. Extracellular vesicle-functionalized bioactive scaffolds for bone regeneration. Asian J Pharm Sci 2024; 19:100945. [PMID: 39483718 PMCID: PMC11525715 DOI: 10.1016/j.ajps.2024.100945] [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: 12/26/2023] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 11/03/2024] Open
Abstract
The clinical need for effective bone regeneration in compromised conditions continues to drive demand for innovative solutions. Among emerging strategies, extracellular vesicles (EVs) have shown promise as an acellular approach for bone regeneration. However, their efficacy is hindered by rapid sequestration and clearance when administered via bolus injection. To address this challenge, EV-functionalized scaffolds have recently been proposed as an alternative delivery strategy to enhance EV retention and subsequent healing efficacy. This review aims to consolidate recent advancements in the development of EV-functionalized scaffolds for augmenting bone regeneration. It explores various sources of EVs and different strategies for integrating them into biomaterials. Furthermore, the mechanisms underlying their therapeutic effects in bone regeneration are elucidated. Current limitations in clinical translation and perspectives on the design of more efficient EVs for improved therapeutic efficacy are also presented. Overall, this review can provide inspiration for the development of novel EV-assisted grafts with superior bone regeneration potential.
Collapse
Affiliation(s)
- Taozhao Yu
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
| | - Irene Shuping Zhao
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| | - Hongguang Pan
- Department of Otolaryngology, Shenzhen Children Hospital, Shenzhen 518034, China
| | - Jianhua Yang
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Yongqiang Deng
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen 518055, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
6
|
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
|
7
|
Xia M, Ding J, Wu S, Yan Z, Wang L, Dong M, Niu W. Milk-derived small extracellular vesicles inhibit the MAPK signaling pathway through CD36 in chronic apical periodontitis. Int J Biol Macromol 2024; 274:133422. [PMID: 38925187 DOI: 10.1016/j.ijbiomac.2024.133422] [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: 05/06/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Small extracellular vesicles derived from milk (Milk-sEVs) have the advantages of easy availability, low cost, low toxicity, and inhibition of inflammation. CD36 mediates inflammation stress in a variety of disease states. The purpose of this study was to investigate the role of Milk-sEVs in inhibiting fibroblast inflammation through CD36 and provide reference data for the treatment of chronic apical periodontitis. RESULTS The addition of Milk-sEVs resulted in decreased expression of inflammation-related factors in L929 cells, and transcriptome sequencing screened for the DEG CD36 in the Milk-sEV treatment group under inflammation. The mouse model of apical periodontitis was successfully established, and CD36 expression increased with the development of inflammation. Transfection of si-CD36 into L929 cells reduced inflammation by inhibiting activation of the MAPK signaling pathway. CONCLUSIONS CD36 expression increased with the development of apical periodontitis. In the setting of LPS-mediated inflammation, Milk-sEVs inhibited activation of the MAPK signaling pathway by decreasing the expression of CD36 in L929 cells and thereby reducing inflammation.
Collapse
Affiliation(s)
- Meng Xia
- School of Stomatology, Dalian Medical University, Liaoning 116044, China
| | - Jiayin Ding
- School of Stomatology, Dalian Medical University, Liaoning 116044, China
| | - Saixuan Wu
- School of Stomatology, Dalian Medical University, Liaoning 116044, China.
| | - Zhengru Yan
- School of Stomatology, Dalian Medical University, Liaoning 116044, China
| | - Lina Wang
- School of Stomatology, Dalian Medical University, Liaoning 116044, China.
| | - Ming Dong
- School of Stomatology, Dalian Medical University, Liaoning 116044, China.
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Liaoning 116044, China.
| |
Collapse
|
8
|
He X, Liu Y, Dai Z, Chen Y, Liu W, Dai H, Hu Y. Yoda1 pretreated BMSC derived exosomes accelerate osteogenesis by activating phospho-ErK signaling via Yoda1-mediated signal transmission. J Nanobiotechnology 2024; 22:407. [PMID: 38987801 PMCID: PMC11234696 DOI: 10.1186/s12951-024-02669-0] [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: 11/27/2023] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Segmental bone defects, arising from factors such as trauma, tumor resection, and congenital malformations, present significant clinical challenges that often necessitate complex reconstruction strategies. Hydrogels loaded with multiple osteogenesis-promoting components have emerged as promising tools for bone defect repair. While the osteogenic potential of the Piezo1 agonist Yoda1 has been demonstrated previously, its hydrophobic nature poses challenges for effective loading onto hydrogel matrices.In this study, we address this challenge by employing Yoda1-pretreated bone marrow-derived mesenchymal stem cell (BMSCs) exosomes (Exo-Yoda1) alongside exosomes derived from BMSCs (Exo-MSC). Comparatively, Exo-Yoda1-treated BMSCs exhibited enhanced osteogenic capabilities compared to both control groups and Exo-MSC-treated counterparts. Notably, Exo-Yoda1-treated cells demonstrated similar functionality to Yoda1 itself. Transcriptome analysis revealed activation of osteogenesis-associated signaling pathways, indicating the potential transduction of Yoda1-mediated signals such as ErK, a finding validated in this study. Furthermore, we successfully integrated Exo-Yoda1 into gelatin methacryloyl (GelMA)/methacrylated sodium alginate (SAMA)/β-tricalcium phosphate (β-TCP) hydrogels. These Exo-Yoda1-loaded hydrogels demonstrated augmented osteogenesis in subcutaneous ectopic osteogenesis nude mice models and in rat skull bone defect model. In conclusion, our study introduces Exo-Yoda1-loaded GELMA/SAMA/β-TCP hydrogels as a promising approach to promoting osteogenesis. This innovative strategy holds significant promise for future widespread clinical applications in the realm of bone defect reconstruction.
Collapse
Affiliation(s)
- Xi He
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, 310002, China
| | - Yanling Liu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhongyu Dai
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Yu Chen
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Wenbin Liu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, 410078, China.
| | - Honglian Dai
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China.
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, 310002, China.
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China.
| |
Collapse
|
9
|
Medel-Martinez A, Redrado-Osta A, Crespo-Barreda A, Sancho-Albero M, Sánchez L, Sebastián V, Pardo M, de la Vieja A, Martín-Duque P. Isolation and Characterization of Milk Exosomes for Use in Advanced Therapies. Biomolecules 2024; 14:810. [PMID: 39062527 PMCID: PMC11275001 DOI: 10.3390/biom14070810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Exosomes are cell-derived extracellular vesicles (EVs) with diameters between 30 and 120 nm. In recent years, several studies have evaluated the therapeutic potential of exosomes derived from different fluids due to their low immunogenicity and high biocompatibility. However, producing exosomes on a large scale is still challenging. One of the fluids from which they could be isolated in large quantities is milk. Moreover, regeneration is a well-known property of milk. The present work seeks to optimize a method for isolating exosomes from bovine and human milk, comparing different storage conditions and different extraction protocols. We found differences in the yield extraction associated with pre-storage milk conditions and observed some differences according to the processing agent. When we removed milk fat globules and added rennet before freezing, we obtained a cleaner final fraction. In summary, we attempted to optimize a rennet-based new milk-exosome isolation method and concluded that pre-treatment, followed by freezing of samples, yielded the best exosome population.
Collapse
Affiliation(s)
- Ana Medel-Martinez
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain (M.S.-A.); (V.S.)
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| | - Ana Redrado-Osta
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain (M.S.-A.); (V.S.)
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| | | | - Maria Sancho-Albero
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain (M.S.-A.); (V.S.)
- Instituto de Nanociencia y Materiales de Aragon (INMA), CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I+D, C/Poeta Mariano Esquillor, s/n, 50018 Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018 Zaragoza, Spain
| | - Lourdes Sánchez
- Instituto Agroalimentario de Aragón, University of Zaragoza, 50018 Zaragoza, Spain;
| | - Víctor Sebastián
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain (M.S.-A.); (V.S.)
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- Instituto de Nanociencia y Materiales de Aragon (INMA), CSIC-Universidad de Zaragoza, Campus Rio Ebro, Edificio I+D, C/Poeta Mariano Esquillor, s/n, 50018 Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Chemical and Environmental Engineering, University of Zaragoza, Campus Rio Ebro, C/María de Luna, 3, 50018 Zaragoza, Spain
| | - María Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), CIBEROBN (ISCIII), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), 15706 Santiago de Compostela, Spain
| | - Antonio de la Vieja
- Endocrine Tumors Unit, Chronic Disease Program (UFIEC), Instituto de Salud Carlos III, 28222 Majadahonda, Spain;
| | - Pilar Martín-Duque
- Instituto de Investigaciones Sanitarias de Aragón (IIS Aragón), 50009 Zaragoza, Spain (M.S.-A.); (V.S.)
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
- Networking Research Center in Biomaterials, Bioengineering and Nanomedicine (CIBERBBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Nanomedicines and Nanotherapies Unit, Departament of Development of Medicines and Advanced Therapies, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
- Surgery Department, Medical School, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
10
|
Chen JG, Zhang EC, Wan YY, Huang TY, Wang YC, Jiang HY. Engineered hsa-miR-455-3p-Abundant Extracellular Vesicles Derived from 3D-Cultured Adipose Mesenchymal Stem Cells for Tissue-Engineering Hyaline Cartilage Regeneration. Adv Healthc Mater 2024; 13:e2304194. [PMID: 38508211 DOI: 10.1002/adhm.202304194] [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: 11/28/2023] [Revised: 03/04/2024] [Indexed: 03/22/2024]
Abstract
Efforts are made to enhance the inherent potential of extracellular vesicles (EVs) by utilizing 3D culture platforms and engineered strategies for functional cargo-loading. Three distinct types of adipose mesenchymal stem cells-derived EVs (ADSCs-EVs) are successfully isolated utilizing 3D culture platforms consisting of porous gelatin methacryloyl (PG), PG combined with sericin methacryloyl (PG/SerMA), or PG combined with chondroitin sulfate methacryloyl (PG/ChSMA). These correspond to PG-EVs, PG/SerMA-EVs, and PG/ChSMA-EVs, respectively. Unique microRNA (miRNA) profiles are observed in each type of ADSCs-EVs. Notably, PG-EVs encapsulate higher levels of hsa-miR-455-3p and deliver more hsa-miR-455-3p to chondrocytes, which results in the activation of the hsa-miR-455-3p/PAK2/Smad2/3 axis and the subsequent hyaline cartilage regeneration. Furthermore, the functionality of PG-EVs is optimized through engineered strategies, including agomir/lentivirus transfection, electroporation, and Exo-Fect transfection. These strategies, referred to as Agomir-EVs, Lentivirus-EVs, Electroporation-EVs, and Exo-Fect-EVs, respectively, are ranked based on their efficacy in encapsulating hsa-miR-455-3p, delivering hsa-miR-455-3p to chondrocytes, and promoting cartilage formation via the hsa-miR-455-3p/PAK2/Smad2/3 axis. Notably, Exo-Fect-EVs exhibit the highest efficiency. Collectively, the 3D culture conditions and engineered strategies have an impact on the miRNA profiles and cartilage regeneration capabilities of ADSCs-EVs. The findings provide valuable insights into the mechanisms underlying the promotion of cartilage regeneration by ADSCs-EVs.
Collapse
Affiliation(s)
- Jian-Guo Chen
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| | - En-Chong Zhang
- Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China
| | - Ying-Ying Wan
- Beijing University of Chinese Medicine, DongFang Hospital, Fengtai District, Beijing, 100078, China
| | - Tian-Yu Huang
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| | - Yu-Chen Wang
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| | - Hai-Yue Jiang
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Shijingshan District, Beijing, 100144, China
| |
Collapse
|
11
|
Wang L, Zhang G, Gao Y, Dai T, Yu J, Liu Y, Bao H, She J, Hou Y, Kong L, Cai B. Extracellular Vesicles Derived from Neutrophils Accelerate Bone Regeneration by Promoting Osteogenic Differentiation of BMSCs. ACS Biomater Sci Eng 2024; 10:3868-3882. [PMID: 38703236 PMCID: PMC11167592 DOI: 10.1021/acsbiomaterials.4c00106] [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: 01/18/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
The reconstruction of bone defects has been associated with severe challenges worldwide. Nowadays, bone marrow mesenchymal stem cell (BMSC)-based cell sheets have rendered this approach a promising way to facilitate osteogenic regeneration in vivo. Extracellular vesicles (EVs) play an essential role in intercellular communication and execution of various biological functions and are often employed as an ideal natural endogenous nanomedicine for restoring the structure and functions of damaged tissues. The perception of polymorphonuclear leukocytes (neutrophils, PMNs) as indiscriminate killer cells is gradually changing, with new evidence suggesting a role for these cells in tissue repair and regeneration, particularly in the context of bone healing. However, the role of EVs derived from PMNs (PMN-EVs) in bone regeneration remains largely unknown, with limited research being conducted on this aspect. In the current study, we investigated the effects of PMN-EVs on BMSCs and the underlying molecular mechanisms as well as the potential application of PMN-EVs in bone regeneration. Toward this end, BMSC-based cell sheets with integrated PMN-EVs (BS@PMN-EVs) were developed for bone defect regeneration. PMN-EVs were found to significantly enhance the proliferation and osteogenic differentiation of BMSCs in vitro. Furthermore, BS@PMN-EVs were found to significantly accelerate bone regeneration in vivo by enhancing the maturation of the newly formed bone in rat calvarial defects; this is likely attributable to the effect of PMN-EVs in promoting the expression of key osteogenic proteins such as SOD2 and GJA1 in BMSCs. In conclusion, our findings demonstrate the crucial role of PMN-EVs in promoting the osteogenic differentiation of BMSCs during bone regeneration. Furthermore, this study proposes a novel strategy for enhancing bone repair and regeneration via the integration of PMN-EVs with BMSC-based cell sheets.
Collapse
Affiliation(s)
- Le Wang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Guanhua Zhang
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral Implants, School
of Stomatology, The Fourth Military Medical
University, Xi’an 710032, China
| | - Ye Gao
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Taiqiang Dai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Jie Yu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Ya Liu
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
- College
of Life Sciences, Northwest University, Xi’an 710069, China
| | - Han Bao
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Jianzhen She
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Yan Hou
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| | - Bolei Cai
- State
Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration,
National Clinical Research Center for Oral Diseases, Shaanxi Clinical
Research Center for Oral Diseases, Department of Oral and Maxillofacial
Surgery, School of Stomatology, The Fourth
Military Medical University, Xi’an 710032, China
| |
Collapse
|
12
|
Prasadani M, Kodithuwakku S, Pennarossa G, Fazeli A, Brevini TAL. Therapeutic Potential of Bovine Milk-Derived Extracellular Vesicles. Int J Mol Sci 2024; 25:5543. [PMID: 38791583 PMCID: PMC11122584 DOI: 10.3390/ijms25105543] [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: 04/16/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Milk is a fundamental component of the human diet, owing to its substantial nutritional content. In addition, milk contains nanoparticles called extracellular vesicles (EVs), which have indicated their potential beneficial roles such as cell-to-cell communication, disease biomarkers, and therapeutics agents. Amidst other types of EVs, milk EVs (MEVs) have their significance due to their high abundance, easy access, and stability in harsh environmental conditions, such as low pH in the gut. There have been plenty of studies conducted to evaluate the therapeutic potential of bovine MEVs over the past few years, and attention has been given to their engineering for drug delivery and targeted therapy. However, there is a gap between the experimental findings available and clinical trials due to the many challenges related to EV isolation, cargo, and the uniformity of the material. This review aims to provide a comprehensive comparison of various techniques for the isolation of MEVs and offers a summary of the therapeutic potential of bovine MEVs described over the last decade, analyzing potential challenges and further applications. Although a number of aspects still need to be further elucidated, the available data point to the role of MEVs as a potential candidate with therapeutics potential, and the supplementation of MEVs would pave the way to understanding their in-depth effects.
Collapse
Affiliation(s)
- Madhusha Prasadani
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia; (M.P.); (S.K.); (A.F.)
| | - Suranga Kodithuwakku
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia; (M.P.); (S.K.); (A.F.)
- Department of Animal Sciences, Faculty of Agriculture, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Georgia Pennarossa
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Center for Stem Cell Research, Università degli Studi di Milano, 26900 Lodi, Italy;
| | - Alireza Fazeli
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51006 Tartu, Estonia; (M.P.); (S.K.); (A.F.)
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2SF, UK
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Center for Stem Cell Research, Università degli Studi di Milano, 26900 Lodi, Italy;
| |
Collapse
|
13
|
Sun M, Zhang H, Liu J, Chen J, Cui Y, Wang S, Zhang X, Yang Z. Extracellular Vesicles: A New Star for Gene Drug Delivery. Int J Nanomedicine 2024; 19:2241-2264. [PMID: 38465204 PMCID: PMC10924919 DOI: 10.2147/ijn.s446224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/20/2024] [Indexed: 03/12/2024] Open
Abstract
Recently, gene therapy has become a subject of considerable research and has been widely evaluated in various disease models. Though it is considered as a stand-alone agent for COVID-19 vaccination, gene therapy is still suffering from the following drawbacks during its translation from the bench to the bedside: the high sensitivity of exogenous nucleic acids to enzymatic degradation; the severe side effects induced either by exogenous nucleic acids or components in the formulation; and the difficulty to cross the barriers before reaching the therapeutic target. Therefore, for the successful application of gene therapy, a safe and reliable transport vector is urgently needed. Extracellular vesicles (EVs) are the ideal candidate for the delivery of gene drugs owing to their low immunogenicity, good biocompatibility and low toxicity. To better understand the properties of EVs and their advantages as gene drug delivery vehicles, this review covers from the origin of EVs to the methods of EVs generation, as well as the common methods of isolation and purification in research, with their pros and cons discussed. Meanwhile, the engineering of EVs for gene drugs is also highlighted. In addition, this paper also presents the progress in the EVs-mediated delivery of microRNAs, small interfering RNAs, messenger RNAs, plasmids, and antisense oligonucleotides. We believe this review will provide a theoretical basis for the development of gene drugs.
Collapse
Affiliation(s)
- Man Sun
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Jiayi Liu
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Jiayi Chen
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Yaxin Cui
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Simiao Wang
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Xiangyu Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310020, People’s Republic of China
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| |
Collapse
|
14
|
Kim NH, Kim J, Lee JY, Bae HA, Kim CY. Application of Milk Exosomes for Musculoskeletal Health: Talking Points in Recent Outcomes. Nutrients 2023; 15:4645. [PMID: 37960298 PMCID: PMC10647311 DOI: 10.3390/nu15214645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Milk is a nutrient-rich food source, and among the various milks, breast milk is a nutrient source provided by mothers to newborns in many mammals. Exosomes are nano-sized membranous extracellular vesicles that play important roles in cell-to-cell communication. Exosomes originate from endogenous synthesis and dietary sources such as milk. Discovered through electron microscopy as floating vesicles, the existence of exosomes in human milk was confirmed owing to a density between 1.10 and 1.18 g/mL in a sucrose gradient corresponding to the known density of exosomes and detection of MHC classes I and II, CD63, CD81, and CD86 on the vesicles. To date, milk exosomes have been used for treating many diseases, including cancers, and are widely proposed as promising carriers for the delivery of chemotherapeutic agents. However, few studies on milk exosomes focus on geriatric health, especially sarcopenia and osteoporosis related to bone and muscle. Therefore, the present study focused on milk exosomes and their cargoes, which are potential candidates for dietary supplements, and when combined with drugs, they can be effective in treating musculoskeletal diseases. In this review, we introduce the basic concepts, including the definition, various sources, and cargoes of milk exosomes, and exosome isolation and characterization methods. Additionally, we review recent literature on the musculoskeletal system and milk exosomes. Since inflammation and oxidative stress underly musculoskeletal disorders, studies reporting the antioxidant and anti-inflammatory properties of milk exosomes are also summarized. Finally, the therapeutic potential of milk exosomes in targeting muscle and bone health is proposed.
Collapse
Affiliation(s)
- Na-Hyung Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea; (N.-H.K.); (J.K.); (J.-Y.L.); (H.-A.B.)
- Department of Food and Nutrition, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Juhae Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea; (N.-H.K.); (J.K.); (J.-Y.L.); (H.-A.B.)
| | - Joo-Yeon Lee
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea; (N.-H.K.); (J.K.); (J.-Y.L.); (H.-A.B.)
- Department of Food and Nutrition, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Hyeon-A Bae
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea; (N.-H.K.); (J.K.); (J.-Y.L.); (H.-A.B.)
- Department of Food and Nutrition, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Choon Young Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea; (N.-H.K.); (J.K.); (J.-Y.L.); (H.-A.B.)
- Department of Food and Nutrition, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| |
Collapse
|
15
|
Zhang Y, Lin Y, He J, Song S, Luo Y, Lu Y, Chen S, Wang Q, Li Y, Ren F, Guo H. Milk-derived small extracellular vesicles: a new perspective on dairy nutrition. Crit Rev Food Sci Nutr 2023; 64:13225-13246. [PMID: 37819268 DOI: 10.1080/10408398.2023.2263573] [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: 10/13/2023]
Abstract
Milk contains bioactive compounds that have multiple essential benefits. Milk-derived small extracellular vesicles (M-sEVs) have emerged as novel bioactive milk components with various beneficial biological functions and broad applications. The M-sEVs from different mammalian sources have similar composition and bioactive functions. The digestive stability and biocompatibility of the M-sEVs provide a good foundation for their physiological functions. Evidence suggests that M-sEVs promote intestinal, immune, bone, neural, liver, and heart health and show therapeutic effects against cancer, indicating their potential for use in functional foods. In addition, M-sEVs can be developed as natural delivery carriers owing to their superior structural characteristics. Further studies are needed to elucidate the relationship between the specific components and functions of M-sEVs, standardize their extraction processes, and refine relevant clinical trials to advance the future applications of M-sEVs. This review summarizes the structure and composition of M-sEVs isolated from different milk sources and discusses several common extraction methods. Since the introduction of M-sEVs for digestion and absorption, studies have been conducted on their biological functions. Furthermore, we outline the theoretical industrial production route, potential application scenarios of M-sEVs, and the future perspectives of M-sEV research.
Collapse
Affiliation(s)
- Yuning Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Sijia Song
- Food Laboratory of Zhongyuan, Luohe, PR China
| | - Yujia Luo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | | | - Qingyu Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
16
|
Liu R, Liu S, Wu S, Xia M, Liu W, Wang L, Dong M, Niu W. Milk-Derived Small Extracellular Vesicles Promote Osteogenic Differentiation and Inhibit Inflammation via microRNA-21. Int J Mol Sci 2023; 24:13873. [PMID: 37762176 PMCID: PMC10531249 DOI: 10.3390/ijms241813873] [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: 08/01/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic apical periodontitis (CAP) is a disease with characteristics of inflammation and bone loss. In this study, our objective was to examine the function of small extracellular vesicles (sEVs) obtained from milk in encouraging osteogenic differentiation and inhibiting inflammation by miR-21 in CAP. The expression of miR-21 was detected using qRT-PCR in human CAP samples. The impact of miR-21 on the process of osteogenic differentiation was investigated using CCK-8, qRT-PCR, immunofluorescence staining, and Western blot analysis. The evaluation of RAW 264.7 cell polarization and the assessment of inflammatory factor expression were conducted through qRT-PCR. The influence of sEVs on MC3T3-E1 cells and RAW 264.7 cells was examined, with a particular emphasis on the involvement of miR-21. In human CAP samples, a decrease in miR-21 expression was observed. MiR-21 increased the expression of osteogenesis-related genes and M2 polarization genes while decreasing the expression of M1 polarization genes and inflammatory cytokines. Treatment with milk-derived sEVs also promoted osteogenesis and M2 polarization while inhibiting M1 polarization and inflammation. Conversely, the addition of miR-21 inhibitors resulted in opposite effects. Our results indicated that sEVs derived from milk had a positive effect on bone formation and activation of anti-inflammatory (M2) macrophages and simultaneously reduced inflammation by regulating miR-21 in CAP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian 116044, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
17
|
Davies OG. Extracellular vesicles: From bone development to regenerative orthopedics. Mol Ther 2023; 31:1251-1274. [PMID: 36869588 PMCID: PMC10188641 DOI: 10.1016/j.ymthe.2023.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Regenerative medicine aims to promote the replacement of tissues lost to damage or disease. While positive outcomes have been observed experimentally, challenges remain in their clinical translation. This has led to growing interest in applying extracellular vesicles (EVs) to augment or even replace existing approaches. Through the engineering of culture environments or direct/indirect manipulation of EVs themselves, multiple avenues have emerged to modulate EV production, targeting, and therapeutic potency. Drives to modulate release using material systems or functionalize implants for improved osseointegration have also led to outcomes that could have real-world impact. The purpose of this review is to highlight advantages in applying EVs for the treatment of skeletal defects, outlining the current state of the art in the field and emphasizing avenues for further investigation. Notably, the review identifies inconsistencies in EV nomenclature and outstanding challenges in defining a reproducible therapeutic dose. Challenges also remain in the scalable manufacture of a therapeutically potent and pure EV product, with a need to address scalable cell sources and optimal culture environments. Addressing these issues will be critical if we are to develop regenerative EV therapies that meet the demands of regulators and can be translated from bench to bedside.
Collapse
Affiliation(s)
- Owen G Davies
- School of Sport, Exercise, and Health Sciences, Loughborough University, Epinal Way, Loughborough, Leicestershire LE11 3TU, UK.
| |
Collapse
|