1
|
Zhu H, Liu F, Liao Y, Li H, Gao K, Liang X, Jiang H, Chen F, Wu J, Wang Q, Wang Y, Shuai X, Yi X. Biomimetic nanostructural materials based on placental amniotic membrane-derived nanofibers for self-healing and anti-adhesion during cesarean section. Biomaterials 2025; 317:123081. [PMID: 39787897 DOI: 10.1016/j.biomaterials.2024.123081] [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: 09/23/2024] [Revised: 12/05/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
Cesarean section (CS) is highly prevalent surgery among females. However, current absorbable anti-adhesion membranes used clinically can partially prevent postoperative adhesions but show limited efficacy in tissue regeneration, leaving post-cesarean women at risk for severe complications including cesarean scar pregnancy, placenta previa, and uterine rupture. Herein, we designed a fully amniotic membrane (AM)-derived biomimetic nanostructural materials (AM-BNMs) as an anti-adhesion barrier, and validated its therapeutic efficacy in a rat CS model. The biomaterial consisted of AM-extracellular matrix (ECM) nanofibers, enriched with hemostatic proteins (collagen, S100A8, S100A9, etc.), carrying AM mesenchymal stem cells (MSCs)-secretome that exhibited significantly elevated levels of pro-regenerative factors (miR-302a-3p, angiogenin, VEGF, etc.) compared to endogenous secretion. The reconstituted AM-BNMs demonstrated synergistic effects at CS wounds, effectively preventing adhesion formation while promoting hemostasis and tissue regeneration. In summary, this readily accessible human-derived biomaterial shows promising potential in preventing adhesion-related complications and enhancing uterine wound healing, thereby promoting female reproductive health.
Collapse
Affiliation(s)
- Honglei Zhu
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Fenghua Liu
- Center for Reproductive Medicine, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Yuru Liao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huayan Li
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kunjie Gao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaomei Liang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Haoyuan Jiang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Feng Chen
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jianwei Wu
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qian Wang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yifeng Wang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xiao Yi
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511462, China.
| |
Collapse
|
2
|
Abbad L, Esteve E, Chatziantoniou C. Advances and challenges in kidney fibrosis therapeutics. Nat Rev Nephrol 2025; 21:314-329. [PMID: 39934355 DOI: 10.1038/s41581-025-00934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Chronic kidney disease (CKD) is a major global health burden that affects more than 10% of the adult population. Current treatments, including dialysis and transplantation, are costly and not curative. Kidney fibrosis, defined as an abnormal accumulation of extracellular matrix in the kidney parenchyma, is a common outcome in CKD, regardless of disease aetiology, and is a major cause of loss of kidney function and kidney failure. For this reason, research efforts have focused on identifying mediators of kidney fibrosis to inform the development of effective anti-fibrotic treatments. Given the prominent role of the transforming growth factor-β (TGFβ) family in fibrosis, efforts have focused on inhibiting TGFβ signalling. Despite hopes raised by the efficacy of this approach in preclinical models, translation into clinical practice has not met expectations. Antihypertensive and antidiabetic drugs slow the decline in kidney function and could slow fibrosis but, owing to the lack of technologies for in vivo renal imaging, their anti-fibrotic effect cannot be truly assessed at present. The emergence of new drugs targeting pro-fibrotic signalling, or enabling cell repair and cell metabolic reprogramming, combined with better stratification of people with CKD and the arrival of nanotechnologies for kidney-specific drug delivery, open up new perspectives for the treatment of this major public health challenge.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Emmanuel Esteve
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France.
| |
Collapse
|
3
|
Wang Z, Zhang C. Nanomaterials for targeted therapy of kidney diseases: Strategies and advances. Mater Today Bio 2025; 31:101534. [PMID: 39990736 PMCID: PMC11846943 DOI: 10.1016/j.mtbio.2025.101534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
The treatment and management of kidney diseases pose a significant global burden. Due to the presence of blood circulation barriers and glomerular filtration barriers, drug therapy for kidney diseases faces challenges such as poor renal targeting, short half-life, and severe systemic side effects, severely hindering therapeutic progress. Therefore, the research and development of kidney-targeted therapeutic agents is of great clinical significance. In recent years, the application of nanotechnology in the field of nephrology has shown potential for revolutionizing the diagnosis and treatment of kidney diseases. Carefully designed nanomaterials can exhibit optimal biological characteristics, influencing various aspects such as circulation, retention, targeting, and excretion. Rationally designing and modifying nanomaterials based on the anatomical structure and pathophysiological environment of the kidney to achieve highly specific kidney-targeted nanomaterials or nanodrug delivery systems is both feasible and promising. Based on the targeted therapy of kidney diseases, this review discusses the advantages and limitations of current nanomedicine in the targeted therapy of kidney diseases, and summarizes the application and challenges of current renal active/passive targeting strategies, in order to further promote the development of kidney-targeted nanomedicine through a preliminary summary of previous studies and future prospects.
Collapse
Affiliation(s)
- Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
4
|
Abdulmonem WA, Ahsan M, Mallick AK, Mohamed AH, Waggiallah HA, Shafie A, Alzahrani HS, Ashour AA, Rab SO, Mirdad MT, Ali HTO. The Role of Exosomal miRNAs in Female Infertility: Therapeutic Potential and Mechanisms of Action. Stem Cell Rev Rep 2025:10.1007/s12015-025-10869-w. [PMID: 40126819 DOI: 10.1007/s12015-025-10869-w] [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] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Reproductive disorders, including preeclampsia (PE), endometriosis, premature ovarian failure (POF), and polycystic ovary syndrome (PCOS), present substantial challenges to women's reproductive health. Exosomes (EXOs) are cell-derived vesicles containing molecules that influence target cells' gene expression and cellular behavior. Among their cargo, microRNAs (miRNAs)-short, non-coding RNAs typically 19-25 nucleotides in length-play a crucial role in post-transcriptional gene regulation and have been extensively studied for their therapeutic potential. miRNAs are considered therapeutic targets because they regulate key cellular pathways such as proliferation, apoptosis, angiogenesis, and tissue repair. This review examines the role of exosomal miRNAs from sources such as mesenchymal stem cells (MSCs), plasma, and amniotic fluid in female reproductive disorders, including PE, POF, PCOS, and endometriosis. We discuss their biological origins, mechanisms of miRNA sorting and packaging, and their therapeutic applications in modulating disease progression. By categorizing miRNAs according to their beneficial or detrimental effects in specific conditions, we aim to simplify the understanding of their roles in female infertility.
Collapse
Affiliation(s)
- Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Marya Ahsan
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 13317, Saudi Arabia
| | - Ayaz Khurram Mallick
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif, 21944, Saudi Arabia
| | - Hassan Swed Alzahrani
- Counseling Healthy Marriage, Jeddah Regional Laboratory, Jeddah First Cluster , Jeddah, Saudi Arabia
| | - Amal Adnan Ashour
- Department of Oral & Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Tarek Mirdad
- Medical Intern MBBS, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hatim T O Ali
- Obstetrics and Gynecology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
5
|
Zhu Y, Yang H, Xue Z, Tang H, Chen X, Liao Y. Mesenchymal stem cells-derived small extracellular vesicles and apoptotic extracellular vesicles for wound healing and skin regeneration: a systematic review and meta-analysis of preclinical studies. J Transl Med 2025; 23:364. [PMID: 40128791 PMCID: PMC11934660 DOI: 10.1186/s12967-024-05744-0] [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: 07/06/2024] [Accepted: 10/07/2024] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Studies examining the therapeutic potential of Mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) in wound healing and skin regeneration have progressed rapidly. Prior to considering clinical translation, a systematic and comprehensive understanding of these experimental details and the overall impact of MSC-EVs on skin regeneration is necessary. METHODS 83 studies were identified in Web of Science, Embase, and PubMed that satisfied a set of prespecified inclusion criteria. A random effects meta-analysis was conducted for wound closure rate, scar width, blood vessel density and collagen deposition. CONCLUSIONS Our findings demonstrate clear potential of MSC-EVs to be developed as therapy for wound healing and skin regeneration both in diabetic and non-diabetic animal models. Moreover, subgroup analyses demonstrated that apoptotic small extracellular vesicles (ApoSEVs) showed better efficacy than apoptotic bodies (ApoBDs) and small extracellular vesicles (sEVs) in wound closure outcome and collagen deposition, while sEVs displayed better than ApoEVs in revascularization. Among frequently used routes of administration, subcutaneous injection displayed a greater improvement to wound closure, collagen deposition and revascularization as compared to dressing/covering. Among easier-access source of MSCs, ADSCs demonstrated the best effect in wound closure rate and collagen deposition, as compared, BMMSCs displayed better in revascularization. Additionally, high heterogeneity observed in collection conditions, separation methods, storage methods, modifications, treatment dose, administration route, and frequency of MSC-EVs underscores the urgent need for standardization in these areas, prior to clinical translation. PROTOCOL REGISTRATION PROSPERO CRD42024499172.
Collapse
Affiliation(s)
- Yufan Zhu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Han Yang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhixin Xue
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Haojing Tang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China
| | - Xihang Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China.
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, P. R. China.
| |
Collapse
|
6
|
Yoo D, Jung SY, Go D, Park JY, You DG, Jung WK, Li Y, Ding J, Park JH, Um W. Functionalized extracellular vesicles of mesenchymal stem cells for regenerative medicine. J Nanobiotechnology 2025; 23:219. [PMID: 40102934 PMCID: PMC11921732 DOI: 10.1186/s12951-025-03300-6] [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: 06/09/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Stem cell-derived extracellular vesicles (EVs) have emerged as a safe and potent alternative to regenerative medicine in recent decades. Furthermore, the adjustment of EV functions has been recently enabled by certain stem cell preconditioning methods, providing an exceptional opportunity to enhance the therapeutic potential or confer additional functions of stem cell-derived EVs. In this review, we discuss the recent progress of functionalized EVs, based on stem cell preconditioning, for treating various organ systems, such as the musculoskeletal system, nervous system, integumentary system, cardiovascular system, renal system, and respiratory system. Additionally, we summarize the expected outcomes of preconditioning methods for stem cells and their EVs. With recent progress, we suggest considerations and future directions for developing personalized medicine based on preconditioned stem cell-derived EVs.
Collapse
Affiliation(s)
- Donghyeon Yoo
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Se Young Jung
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dabin Go
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Ji Yeong Park
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dong Gil You
- Department of Chemical Engineering & Biotechnology, Tech University of Korea, Siheung, 15073, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| | - Yuce Li
- College of Life Science and Health, Wuhan University of Science and Technology (WUST), Wuhan, 430065, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
7
|
Ivanova A, Chalupska R, Louro AF, Firth M, González-King Garibotti H, Hultin L, Kohl F, Lázaro-Ibáñez E, Lindgren J, Musa G, Oude Blenke E, Silva AM, Szeponik L, Taylor A, Viken I, Wang X, Jennbacken K, Wiseman J, Dekker N. Barcoded Hybrids of Extracellular Vesicles and Lipid Nanoparticles for Multiplexed Analysis of Tissue Distribution. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407850. [PMID: 39823165 PMCID: PMC11904941 DOI: 10.1002/advs.202407850] [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: 07/10/2024] [Revised: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Targeted delivery of therapeutic agents is a persistent challenge in modern medicine. Recent efforts in this area have highlighted the utility of extracellular vesicles (EVs) as drug carriers, given that they naturally occur in bloodstream and tissues, and can be loaded with a wide range of therapeutic molecules. However, biodistribution and tissue tropism of EVs remain difficult to study systematically. Here, a multiplexed approach is developed for simultaneous tracking of EVs from various cell lines within a single in vivo experiment. EVs are used from 16 different cell lines, and through controlled fusion with lipid nanoparticles (LNPs) carrying single-stranded DNA barcodes, uniquely barcoded hybrid EV particle (hEV) library is generated. These hEVs are combined for a multiplexed in vivo biodistribution profiling in mice, and discovered that HAP1-derived hEVs demonstrated lung tropism, suggesting that these hEVs may be used for targeted drug delivery into lung tissue. To examine this possibility further, it is shown that HAP1 hEV loaded with Cre mRNA displayed functional delivery to the lungs. Overall, the barcoded hEV technology enables rapid profiling of biodistribution across EV cell sources, which is poised to improve throughput and extent of EV studies, while reducing the number of animals required for research.
Collapse
Affiliation(s)
- Alena Ivanova
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Renata Chalupska
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Ana Filipa Louro
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Hernán González-King Garibotti
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Leif Hultin
- Clinical Pharmacology and Safety Science, Imaging and Data Analytics BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Franziska Kohl
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 1, Solna, Stockholm, 171 77, Sweden
| | - Elisa Lázaro-Ibáñez
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Julia Lindgren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Gentian Musa
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Erik Oude Blenke
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Louis Szeponik
- Clinical Pharmacology and Safety Science, Imaging and Data Analytics BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Agnes Taylor
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Ida Viken
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Xiaoqin Wang
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - John Wiseman
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| |
Collapse
|
8
|
Garcia‐Aponte OF, Kahlenberg S, Kouroupis D, Egger D, Kasper C. Effects of Hydrogels on Mesenchymal Stem/Stromal Cells Paracrine Activity and Extracellular Vesicles Production. J Extracell Vesicles 2025; 14:e70057. [PMID: 40091440 PMCID: PMC11911545 DOI: 10.1002/jev2.70057] [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: 08/07/2024] [Revised: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a valuable source of paracrine factors, as they have a remarkable secretory capacity, and there is a sizeable knowledge base to develop industrial and clinical production protocols. Promising cell-free approaches for tissue regeneration and immunomodulation are driving research towards secretome applications, among which extracellular vesicles (EVs) are steadily gaining attention. However, the manufacturing and application of EVs is limited by insufficient yields, knowledge gaps, and low standardization. Facing these limitations, hydrogels represent a versatile three-dimensional (3D) culture platform that can incorporate extracellular matrix (ECM) components to mimic the natural stem cell environment in vitro; via these niche-mimicking properties, hydrogels can regulate MSCs' morphology, adhesion, proliferation, differentiation and secretion capacities. However, the impact of the hydrogel's architectural, biochemical and biomechanical properties on the production of EVs remains poorly understood, as the field is still in its infancy and the interdependency of culture parameters compromises the comparability of the studies. Therefore, this review summarizes and discusses the reported effects of hydrogel encapsulation and culture on the secretion of MSC-EVs. Considering the effects of cell-material interactions on the overall paracrine activity of MSCs, we identify persistent challenges from low standardization and process control, and outline future paths of research, such as the synergic use of hydrogels and bioreactors to enhance MSC-EV generation.
Collapse
Affiliation(s)
- Oscar Fabian Garcia‐Aponte
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Simon Kahlenberg
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Diabetes Research Institute & Cell Transplant Center, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Dominik Egger
- Institute of Cell Biology and BiophysicsLeibniz University HannoverHannoverGermany
| | - Cornelia Kasper
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
9
|
Xu T, Peng Y, Xu Y, Zhu J, Yang Q, Liu Y, Yang H. Exploring the therapeutic potential of small extracellular vesicles derived from induced pluripotent stem cell in periodontal regeneration. J Oral Biosci 2025; 67:100621. [PMID: 39892783 DOI: 10.1016/j.job.2025.100621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVES To investigate the role of small extracellular vesicles derived from induced pluripotent stem cells (iPSC-sEVs) in periodontal tissue regeneration, elucidate their potential molecular mechanisms, and provide theoretical guidance for the clinical application of iPSC-sEVs as a cell-free therapeutic strategy for periodontal tissue regeneration. METHODS We investigated the effects of iPSC-sEVs on the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells (PDLSCs) in vitro. The regenerative potential of iPSC-sEVs was evaluated in vivo, using a periodontal defect model. Bulk RNA sequencing was performed to elucidate the underlying molecular mechanisms. RESULTS iPSC-sEVs were isolated, characterized, and systemically evaluated for regenerative potential. The results revealed that treatment with iPSC-sEVs significantly enhanced the proliferation, migration, and osteogenic differentiation of PDLSCs. In situ treatment with iPSC-sEVs loaded onto collagen sponges was performed in a rat model of periodontal defects. Micro-CT and histological analyses indicated that iPSC-sEV treatment markedly promoted alveolar bone repair and periodontal ligament regeneration. Mechanistically, the analysis of bulk RNA sequencing data coupled with experimental validation revealed that iPSC-sEV treatment significantly activated the mitogen-activated protein kinase (MAPK) signaling pathway in PDLSCs. Further investigation showed that the inhibition of this pathway completely abolished the proliferative effects of iPSC-sEVs on PDLSCs. CONCLUSIONS iPSC-sEVs promote PDLSC proliferation through MAPK signaling pathway activation, while also enhancing PDLSC migratory and osteogenic differentiation capacities, facilitates the repair and regeneration of damaged periodontal tissue and presents a potential novel therapeutic strategy for clinical periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tingting Xu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Yi Peng
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Yanan Xu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Jing Zhu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Qiao Yang
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Periodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Yali Liu
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China; Department of Prosthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
| |
Collapse
|
10
|
Chen X, Tian B, Wang Y, Zheng J, Kang X. Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review). Int J Mol Med 2025; 55:43. [PMID: 39791222 PMCID: PMC11759586 DOI: 10.3892/ijmm.2025.5484] [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: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Exosomes are integral to the pathophysiology of osteoarthritis (OA) due to their roles in mediating intercellular communication and regulating inflammatory processes. Exosomes are integral to the transport of bioactive molecules, such as proteins, lipids and nucleic acids, which can influence chondrocyte behavior and joint homeostasis. Given their properties of regeneration and ability to target damaged tissues, exosomes represent a promising therapeutic avenue for OA treatment. Exosomes have potential in promoting cartilage repair, reducing inflammation and improving overall joint function. However, several challenges remain, including the need for standardized isolation and characterization methods, variability in exosomal content, and regulatory hurdles. The present review aims to provide a comprehensive overview of the current understanding of exosome mechanisms in OA and their therapeutic potential, while also addressing the ongoing challenges faced in translating these findings into clinical practice. By consolidating existing research, the present review aims to pave the way for future studies aimed at optimizing exosome‑based therapies for effective OA management.
Collapse
Affiliation(s)
| | | | | | - Jiang Zheng
- Department of Joint Surgery, Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| | - Xin Kang
- Department of Joint Surgery, Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| |
Collapse
|
11
|
Tomasello L, Biondo M, Biscari G, Di Rosa L, Palumbo FS, Fiorica C, Pitarresi G, Vasto S, Pizzolanti G, Arnaldi G. Amine-Functionalized Gellan Gum-Based Hydrogel Loaded with Adipose Stem Cell-Derived Small Extracellular Vesicles: An In Vitro Proof of Concept for Enhancing Diabetic Foot Ulcer Healing. Gels 2025; 11:119. [PMID: 39996662 PMCID: PMC11854167 DOI: 10.3390/gels11020119] [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: 12/31/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetic foot ulcers (DFUs) are chronic wounds and a common complication of diabetes. A promising strategy in the treatment of DFUs involves the use of stem cell derivatives, such as small extracellular vesicles (sEVs), which can enhance cell proliferation and reduce inflammation while avoiding immunogenic responses. In this study, we evaluated the ability of adipose mesenchymal stem cell- (ASC)-derived sEVs to enhance the proliferation of human fibroblasts, which play a crucial role in wound regenerative processes. To mimic the inflammatory environment of DFUs, fibroblasts were cultured into the gellan gum (GG) modified with ethylenediamine (EDA) hydrogel scaffolds loaded with ASC-derived sEVs, under pro-inflammatory cytokines. Our comparative analysis demonstrated that sEVs loaded in GG-EDA hydrogel improved fibroblast viability in pro-inflamed conditions while retaining the anti-inflammatory and immunomodulatory properties of their cells of origin. By modulating the gene expression profile of fibroblasts to promote cell proliferation, wound healing and re-epithelialization, our system presents a promising therapeutic strategy for DFU healing.
Collapse
Affiliation(s)
- Laura Tomasello
- Laboratory of Endocrinology and Regenerative Medicine “Aldo Galluzzo”, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, 90127 Palermo, Italy; (G.P.); (G.A.)
| | - Mattia Biondo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Giuseppina Biscari
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Luigi Di Rosa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Fabio Salvatore Palumbo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Calogero Fiorica
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Giovanna Pitarresi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Sonya Vasto
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy; (M.B.); (G.B.); (L.D.R.); (F.S.P.); (G.P.); (S.V.)
| | - Giuseppe Pizzolanti
- Laboratory of Endocrinology and Regenerative Medicine “Aldo Galluzzo”, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, 90127 Palermo, Italy; (G.P.); (G.A.)
- Advanced Technologies Network (ATeN) Center, University of Palermo, 90128 Palermo, Italy
| | - Giorgio Arnaldi
- Laboratory of Endocrinology and Regenerative Medicine “Aldo Galluzzo”, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, 90127 Palermo, Italy; (G.P.); (G.A.)
| |
Collapse
|
12
|
Wen X, Hao Z, Yin H, Min J, Wang X, Sun S, Ruan G. Engineered Extracellular Vesicles as a New Class of Nanomedicine. CHEM & BIO ENGINEERING 2025; 2:3-22. [PMID: 39975802 PMCID: PMC11835263 DOI: 10.1021/cbe.4c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 02/21/2025]
Abstract
Extracellular vesicles (EVs) are secreted from biological cells and contain many molecules with diagnostic values or therapeutic functions. There has been great interest in academic and industrial communities to utilize EVs as tools for diagnosis or therapeutics. In addition, EVs can also serve as delivery vehicles for therapeutic molecules. An indicator of the enormous interest in EVs is the large number of review articles published on EVs, with the focus ranging from their biology to their applications. An emerging trend in EV research is to produce and utilize "engineered EVs", which are essentially the enhanced version of EVs. EV engineering can be conducted by cell culture condition control, genetic engineering, or chemical engineering. Given their nanometer-scale sizes and therapeutic potentials, engineered EVs are an emerging class of nanomedicines. So far, an overwhelming majority of the research on engineered EVs is preclinical studies; there are only a very small number of reported clinical trials. This Review focuses on engineered EVs, with a more specific focus being their applications in therapeutics. The various approaches to producing engineered EVs and their applications in various diseases are reviewed. Furthermore, in vivo imaging of EVs, the mechanistic understandings, and the clinical translation aspects are discussed. The discussion is primarily on preclinical studies while briefly mentioning the clinical trials. With continued interdisciplinary research efforts from biologists, pharmacists, physicians, bioengineers, and chemical engineers, engineered EVs could become a powerful solution for many major diseases such as neurological, immunological, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaowei Wen
- Institute
of Analytical Chemistry and Instrument for Life Science, The Key Laboratory
of Biomedical Information Engineering of Ministry of Education, School
of Life Science and Technology, Xi’an
Jiaotong University, Xi’an, China 710049
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Zerun Hao
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Haofan Yin
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Jie Min
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Xueying Wang
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Sihan Sun
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Gang Ruan
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| |
Collapse
|
13
|
Cai Z, Wang Y, Hu S, Yuan Q, Liu J, Luo C, Jiang L, Huang Y. The efficacy of platelet-derived extracellular vesicles in the treatment of diabetic wounds: a systematic review and meta-analysis of animal studies. Arch Dermatol Res 2025; 317:244. [PMID: 39812853 DOI: 10.1007/s00403-024-03781-9] [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: 11/07/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025]
Abstract
Platelet-derived extracellular vesicles (PEVs) are rich in growth factors and have significant potential for facilitating tissue repair and regeneration. Therefore, we conducted this meta-analysis to assess the efficacy of PEVs in treating diabetic wounds. To assess the efficacy and safety of PEVs in treating diabetic wounds, we conducted a systematic review of several databases and performed a meta-analysis using a random effects model. Nine studies (n = 128 animals) meeting the inclusion criteria for this review were identified. The pooled analysis revealed that compared to the control group, wounds treated with PEVs had a higher healing rate (SMD = 4.43, 95% CI = 2.85-6.01, P < 0.00001). In subgroup analysis, PEVs combined with hydrogel showed better efficacy than PEVs alone (SMD = 7.96, 95% CI = 5.05-10.87, P < 0.00001). Additionally, the PEVs treatment group outperformed the control group in other outcomes, such as vessel density and number, re-epithelialization rate, and collagen deposition. PEVs have the potential to promote angiogenesis at diabetic wound sites and alleviate inflammatory responses, ultimately aiding in wound healing, especially when combined with hydrogels or other medications.
Collapse
Affiliation(s)
- Zhi Cai
- Department of Transfusion, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, Luzhou, People's Republic of China
| | - Yuhan Wang
- Department of Clinical Laboratory, Longmatan District People's Hospital of Luzhou, Luzhou, People's Republic of China
| | - Shan Hu
- Department of Transfusion, Guanghan People's Hospital, Deyang, People's Republic of China
| | - Qiong Yuan
- Department of Transfusion, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Jusong Liu
- Department of Transfusion, Zigong First People's Hospital, Zigong, People's Republic of China
| | - Chengcen Luo
- Department of Transfusion, Zigong Fourth People's Hospital, Zigong, People's Republic of China
| | - Ling Jiang
- Department of Transfusion, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, Luzhou, People's Republic of China.
- Department of Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China.
| | - Yuanshuai Huang
- Department of Transfusion, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, Luzhou, People's Republic of China.
- Department of Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People's Republic of China.
| |
Collapse
|
14
|
Emami A, Arabpour Z, Izadi E. Extracellular vesicles: essential agents in critical bone defect repair and therapeutic enhancement. Mol Biol Rep 2025; 52:113. [PMID: 39798011 DOI: 10.1007/s11033-024-10209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025]
Abstract
Bone serves as a fundamental structural component in the body, playing pivotal roles in support, protection, mineral supply, and hormonal regulation. However, critical-sized bone injuries have become increasingly prevalent, necessitating extensive medical interventions due to limitations in the body's capacity for self-repair. Traditional approaches, such as autografts, allografts, and xenografts, have yielded unsatisfactory results. Stem cell therapy emerges as a promising avenue, but challenges like immune rejection and low cell survival rates hinder its widespread clinical implementation. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have garnered attention for their regenerative capabilities, which surpass those of MSCs themselves. EVs offer advantages such as reduced immunogenicity, enhanced stability, and simplified storage, positioning them as a promising tool in stem cell-based therapies. This review explores the potential of EV-based therapy in bone tissue regeneration, delving into their biological characteristics, communication mechanisms, and preclinical applications across various physiological and pathological conditions. The mechanisms underlying EV-mediated bone regeneration, including angiogenesis, osteoblast proliferation, mineralization, and immunomodulation, are elucidated. Preclinical studies demonstrate the efficacy of EVs in promoting bone repair and neovascularization, even in pathological conditions like osteoporosis. EVs hold promise as a potential alternative for regenerating bone tissue, particularly in the context of critical-sized bone defects, offering new avenues for effective bone defect repair and management.
Collapse
Affiliation(s)
- Asrin Emami
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zohreh Arabpour
- Department of Ophthalmology and Visual Science and University of Illinois, Chicago, IL, 60612, USA
| | - Elaheh Izadi
- Pediatric Cell, and Gene Therapy Research Center Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Zhou L, Cai W, Zhang Y, Zhong W, He P, Ren J, Gao X. Therapeutic effect of mesenchymal stem cell-derived exosome therapy for periodontal regeneration: a systematic review and meta-analysis of preclinical trials. J Orthop Surg Res 2025; 20:27. [PMID: 39780243 PMCID: PMC11715287 DOI: 10.1186/s13018-024-05403-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND To assess the therapeutic effects of mesenchymal stem cell (MSC)-derived exosome therapy on periodontal regeneration and identify treatment factors associated with enhanced periodontal regeneration in recent preclinical studies. METHODS Searches were conducted in PubMed, Cochrane Library, EMBASE, and Web of Science databases until October 10, 2024. A risk of bias (ROB) assessment was performed using the SYRCLE tool. Osteogenic-related parameters were used as the primary outcome measures. RESULTS In total, 1360 articles were identified, of which 17 preclinical studies were based on MSC-derived exosome therapy, and they demonstrated a beneficial effect on BV/TV (SMD = 13.99; 95% Cl = 10.50, 17.48; p < 0.00001), CEJ-ABC (SMD = -0.22; 95% Cl = -0.31, -0.13; p < 0.00001), BMD (SMD = 0.29; 95% Cl = 0.14, 0.45; p = 0.0002), and Tp.Sp (SMD = -0.08; 95% Cl= -0.15, -0.02; p = 0.02) compared with the control group. However, no significant differences were observed in Tp.Th (SMD = 0.03; 95% CI = 0.00, 0.07; p = 0.09) between the exosome-treated group and control group. Additionally, subgroup analysis indicated that preconditioned exosomes (p = 0.03) significantly improved BV/TV. In contrast, there were no significant differences in the enhancement of BV/TV with respect to the application method (p = 0.29), application frequency (p = 0.10), treatment duration (p = 0.15), or source of MSCs (p = 0.31). CONCLUSIONS MSC-derived exosomes show great promise for enhancing the quality of periodontal regeneration. However, more standardized and robust trials are needed to reduce heterogeneity and bias across studies and to confirm the therapeutic parameters associated with the enhancement of periodontal regeneration by MSC-derived exosomes. REGISTRATION CRD42024546236.
Collapse
Affiliation(s)
- Liping Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Wenjia Cai
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Yuhan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China
| | - Ping He
- Department of Stomatology, Dazhou Central Hospital, Dazhou, 635000, China.
| | - Jingsong Ren
- Department of Stomatology, Dazhou Central Hospital, Dazhou, 635000, China.
| | - Xiang Gao
- College of Stomatology, Chongqing Medical University, Chongqing, 401147 , China.
- Chongqing Key Laboratory of Oral Diseases , Chongqing, 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147 , China.
| |
Collapse
|
16
|
Kishta MS, Khamis A, Am H, Elshaar AH, Gül D. Exploring the tumor-suppressive role of miRNA-200c in head and neck squamous cell carcinoma: Potential and mechanisms of exosome-mediated delivery for therapeutic applications. Transl Oncol 2025; 51:102216. [PMID: 39615277 DOI: 10.1016/j.tranon.2024.102216] [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: 09/05/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a challenging malignancy due to its high rates of recurrence, metastasis, and resistance to conventional therapies. microRNA-200c (miRNA-200c) has emerged as a critical tumor suppressor in HNSCC, with the potential to inhibit epithelial-mesenchymal transition (EMT), which is considered as a key process in cancer metastasis and progression. Interestingly, there are also controversial findings in HNSCC characterizing miRNA-200c as oncogenic factor. This review article provides a comprehensive overview of the current understanding of miRNA-200c's general role in cancer, and particularly in HNSCC, highlighting its mechanisms of action, including the regulation of EMT and other oncogenic pathways. Additionally, the review explores the innovative approach of exosome-mediated delivery of miRNA-200c as a therapeutic strategy. Exosomes, as natural nanocarriers, offer a promising vehicle for the targeted delivery of miRNA-200c to tumor cells, potentially overcoming the limitations of traditional delivery methods and enhancing therapeutic efficacy. The review also discusses the challenges and future directions in the clinical application of miRNA-200c, particularly focusing on its potential to improve outcomes for HNSCC patients. This article seeks to provide valuable insights for researchers and clinicians working towards innovative treatments for this aggressive cancer type.
Collapse
Affiliation(s)
- Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, 33 El Bohouth St., Dokki, 12622 Cairo, Egypt.
| | - Aya Khamis
- Maxillofacial and Oral Surgery, University Medical Center, 55131 Mainz, Germany; Oral Pathology Department, Faculty of Dentistry, Alexandria University, 5372066 Alexandria, Egypt
| | - Hafez Am
- Medical Biochemistry Department Faculty of medicine KafrElSheikh University, Kafr El-Sheikh, Egypt
| | | | - Désirée Gül
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany.
| |
Collapse
|
17
|
Wu Z, He L, Yan L, Tan B, Ma L, He G, Dai Z, Sun R, Li C. Hydrogels Treat Atopic Dermatitis by Transporting Marine-Derived miR-100-5p-Abundant Extracellular Vesicles. ACS Biomater Sci Eng 2024; 10:7667-7682. [PMID: 39585960 DOI: 10.1021/acsbiomaterials.4c01649] [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: 11/27/2024]
Abstract
Atopic dermatitis (AD) is a prevalent skin disorder worldwide. However, many AD medications are unsuitable for long-term use due to low therapeutic efficacy and side effects. Extracellular vesicles (EVs) extracted from Pinctada martensii mucus have demonstrated therapeutic efficacy in AD. It is hypothesized that EVs may exert their activity on mammalian cells through their specific contents. In this study, we analyzed the results of miRNA sequencing of the EVs and investigated the potency of highly expressed miR-100-5p in treating AD. To enhance the therapeutic efficiency of the EVs in AD, we developed oxidized sodium alginate (OSA)-carboxymethyl chitosan (CMCS) self-cross-linked hydrogels as a vehicle to deliver the EVs to BALB/c mice with dermatitis. The miR-100-5p in EVs exhibited a favorable anti-inflammatory function, while the hydrogels provided enhanced skin residency. Additionally, its efficacy in inflammation inhibition and collagen synthesis was demonstrated in in vivo experiments. Mechanistically, miR-100-5p in EVs exerted anti-inflammatory effects by inhibiting the expression of FOXO3, consequently suppressing the activation of the downstream NLRP3 signaling pathway. This study underscores the significance of utilizing OSA-CMCS hydrogels as a vehicle for delivering miR-100-5p in P. martensii mucus-derived EVs for the treatment of AD.
Collapse
Affiliation(s)
- Zijie Wu
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
| | - Lei He
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
| | - Linhong Yan
- Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| | - Baoyi Tan
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
| | - Lihua Ma
- Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| | - Guoli He
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhenqing Dai
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| | - Ruikun Sun
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| | - Chengyong Li
- School of Chemistry and Environment, Analytical and Testing Center, Guangdong Provincial Key Laboratory of Intelligent Equipment for South China Sea Marine Ranching, Guangdong Provincial Observation and Research Station for Tropical Ocean Environment in Western Coastal Water, Guangdong Ocean University, Zhanjiang 524088, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen 518108, China
| |
Collapse
|
18
|
Sun X, Ding H, Li X, Wu Y, Huang X. Disulfiram-loaded nanovesicles hydrogel promotes healing of diabetic wound. J Transl Med 2024; 22:1066. [PMID: 39593097 PMCID: PMC11600750 DOI: 10.1186/s12967-024-05875-4] [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: 08/30/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Traditional methods for treating diabetic wounds are limited in effectiveness because of their long healing times, the risk of immune rejection, and susceptibility to infection. Suppressing neutrophil extracellular traps (NETs) is an effective strategy for reducing persistent inflammation in diabetic wounds. Although disulfiram (DSF) can inhibit the significant increase of NETs in diabetic wounds, oral DSF suffers from rapid and harmful metabolism in the liver. To address these challenges, we developed a nanomedicine formulation in which DSF was incorporated into the hydrogel. METHODS In this study, we developed a DSF-laden sodium alginate hydrogel wound dressing, DEP@SA, and characterized its composition, properties, and performance. We examined the effects of DEP@SA on inflammatory phase-related markers such as NETs and their pathway proteins, inflammatory factors, and macrophage phenotypes in a high-glucose environment in vivo and in vitro. In addition, the effects of DEP@SA on tissue regenerative capacity such as epidermal proliferative migration and angiogenesis, were also assessed. RESULTS The results showed that by utilizing extracellular vesicles as a drug delivery system, we effectively mitigated the degradation of DSF via direct contact with aqueous solutions and ensured the stability of DSF@SA, which could then be applied to diabetic wounds. The inflammatory phase-related indicators revealed that DSF@SA effectively reduced inflammation levels, decreased NETs formation, suppressed the Caspase-1/GSDMD pathway in neutrophils, and promoted the polarization of M2 macrophages. Moreover, the hydrogel accelerated wound healing by promoting angiogenesis and re-epithelialization, thereby shortening the diabetic wound healing time. CONCLUSIONS This study confirmed that the DSF@SA composite dressing has the potential to enhance diabetic wound repair and offers a novel approach for drug reutilization.
Collapse
Affiliation(s)
- Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Hanxi Ding
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China.
| |
Collapse
|
19
|
Zhang Y, Wu D, Zhou C, Bai M, Wan Y, Zheng Q, Fan Z, Wang X, Yang C. Engineered extracellular vesicles for tissue repair and regeneration. BURNS & TRAUMA 2024; 12:tkae062. [PMID: 39439545 PMCID: PMC11495891 DOI: 10.1093/burnst/tkae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are heterogeneous membrane-like vesicles secreted by living cells that are involved in many physiological and pathological processes and act as intermediaries of intercellular communication and molecular transfer. Recent studies have shown that EVs from specific sources regulate tissue repair and regeneration by delivering proteins, lipids, and nucleic acids to target cells as signaling molecules. Nanotechnology breakthroughs have facilitated the development and exploration of engineered EVs for tissue repair. Enhancements through gene editing, surface modification, and content modification have further improved their therapeutic efficacy. This review summarizes the potential of EVs in tissue repair and regeneration, their mechanisms of action, and their research progress in regenerative medicine. This review highlights their design logic through typical examples and explores the development prospects of EVs in tissue repair. The aim of this review is to provide new insights into the design of EVs for tissue repair and regeneration applications, thereby expanding their use in regenerative medicine.
Collapse
Affiliation(s)
- Yan Zhang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
- School of Public Health, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Dan Wu
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Chen Zhou
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, No. 3025 Shennan Middle Road, Futian District, Shenzhen, China
| | - Muran Bai
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Yucheng Wan
- Hospital of Stomatology, Zunyi Medical University, No. 89, Wujiang East Road, Xinpu New District, Zunyi City, Guizhou Province, China
| | - Qing Zheng
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Zhijin Fan
- Institute for Engineering Medicine, Kunming Medical University, No. 1168 Chunrong West Road, Yuhua Street, Chenggong District, Kunming City, Yunnan Province China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, No.81 Meishan Road, Shushan District, Hefei 230032, China
| | - Chun Yang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| |
Collapse
|
20
|
Wang G, Li Q, Liu S, Li M, Liu B, Zhao T, Liu B, Chen Z. An injectable decellularized extracellular matrix hydrogel with cortical neuron-derived exosomes enhances tissue repair following traumatic spinal cord injury. Mater Today Bio 2024; 28:101250. [PMID: 39318371 PMCID: PMC11421349 DOI: 10.1016/j.mtbio.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/11/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Traumatic spinal cord injury (SCI), known for its limited intrinsic regeneration capacity, often results in considerable neurological impairment. Studies suggest that therapeutic techniques utilizing exosomes (Exo) to promote tissue regeneration and modulate immune responses are promising for SCI treatment. However, combining exosome therapy with biomaterials for SCI treatment is not very effective. This study developed an adhesive hydrogel using exosomes secreted by cortical neurons derived from human induced pluripotent stem cells (iPSCs) and decellularized extracellular matrix (dECM) from human umbilical cord mesenchymal stem cells (hUCMSCs) to enhance motor function recovery post-SCI. In vitro assessments demonstrated the excellent cytocompatibility of the dECM hydrogel. Additionally, the Exo-dECM hydrogel facilitated the polarization of early M2 macrophages, reduced neuronal apoptosis, and established a pro-regenerative microenvironment in a rodent SCI model. Subsequent analyses revealed significant activation of endogenous neural stem cells and promotion of axon regeneration and remyelination at eight weeks post-surgery. The Exo-dECM hydrogel also promoted the functional recovery and preservation of urinary tissue in SCI-afflicted rats. These findings highlighted that the Exo-dECM hydrogel is a promising therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Gang Wang
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Qian Li
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Sumei Liu
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mo Li
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Baoguo Liu
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Tianyao Zhao
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Bochao Liu
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| |
Collapse
|
21
|
Yi S, Kim J, Kim MJ, Yae CG, Kim KH, Kim HK. Development of human amniotic epithelial cell-derived extracellular vesicles as cell-free therapy for dry eye disease. Ocul Surf 2024; 34:370-380. [PMID: 39332677 DOI: 10.1016/j.jtos.2024.09.006] [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: 01/30/2024] [Revised: 08/16/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
PURPOSE This study aimed to investigate the therapeutic potential of extracellular vesicles (EVs) derived from human amniotic epithelial cells (hAEC-EVs) for Dry Eye Disease (DED) treatment. METHODS Highly purified EVs were isolated from the culture supernatants of hAECs, which obtained from term placenta and characterized. Proteomic contents were analyzed for assessing its biological function related to the therapeutic potentials for DED. Subsequently, we examined the therapeutic efficacy of hAEC-EVs on human corneal epithelial cells exposed to hyperosmotic stress and in an experimental DED mouse model induced by desiccation stress. RESULTS Proteomic analysis of hAEC-EVs revealed proteins linked to cell proliferation and anti-inflammatory responses. We demonstrated efficient uptake of hAEC-EVs by ocular surface cells. Under DED conditions, EV treatment increased corneal epithelial cell proliferation and migration, and concurrently reducing inflammatory cytokines. In the DED mouse model, hAEC-EVs showed significant improvements in corneal staining score, tear secretion, corneal irregularity, and conjunctival goblet cell density. Additionally, hAEC-EVs exhibited a mitigating effect on ocular surface inflammation induced by desiccation. CONCLUSIONS These findings suggest that hAEC-EVs hold potential as a cell-free therapy for corneal epithelial defects and ocular surface diseases, presenting a promising treatment option for DED.
Collapse
Affiliation(s)
- Soojin Yi
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, Republic of Korea; Bio-Medical Institute, Kyungpook National University Hospital, Jung-gu, Daegu, Republic of Korea; Department of Biomedical Science, The Graduate School, Kyungpook National University, Jung-gu, Daegu, Republic of Korea
| | - Jeongho Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, Republic of Korea; Bio-Medical Institute, Kyungpook National University Hospital, Jung-gu, Daegu, Republic of Korea
| | - Mi Ju Kim
- Department of Obstetrics and Gynecology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, Republic of Korea
| | - Che Gyem Yae
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, Republic of Korea
| | - Ki Hean Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea.
| | - Hong Kyun Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Jung-gu, Daegu, Republic of Korea; Bio-Medical Institute, Kyungpook National University Hospital, Jung-gu, Daegu, Republic of Korea.
| |
Collapse
|
22
|
Lee S, Jung SY, Yoo D, Go D, Park JY, Lee JM, Um W. Alternatives of mesenchymal stem cell-derived exosomes as potential therapeutic platforms. Front Bioeng Biotechnol 2024; 12:1478517. [PMID: 39315312 PMCID: PMC11417005 DOI: 10.3389/fbioe.2024.1478517] [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: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
With outstanding therapeutic potential in the tissue regeneration and anti-inflammation, mesenchymal stem cell-derived exosomes (MSC-EXOs) have emerged as a prominent therapeutic in recent. However, poor production yield and reproducibility have remained as significant challenges of their practical applications. To surmount these challenges, various alternative materials with stem cell-like functions, have been recently investigated, however, there has been no comprehensive analysis in these alternatives so far. Here, we discuss the recent progress of alternatives of MSC-EXOs, including exosomes and exosome-like nanovesicles from various biological sources such as plants, milk, microbes, and body fluids. Moreover, we extensively compare each alternative by summarizing their unique functions and mode of actions to suggest the expected therapeutic target and future directions for developing alternatives for MSC-EXOs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jong Min Lee
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
23
|
Tam S, Wear D, Morrone CD, Yu WH. The complexity of extracellular vesicles: Bridging the gap between cellular communication and neuropathology. J Neurochem 2024; 168:2391-2422. [PMID: 38650384 DOI: 10.1111/jnc.16108] [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: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
Brain-derived extracellular vesicles (EVs) serve a prominent role in maintaining homeostasis and contributing to pathology in health and disease. This review establishes a crucial link between physiological processes leading to EV biogenesis and their impacts on disease. EVs are involved in the clearance and transport of proteins and nucleic acids, responding to changes in cellular processes associated with neurodegeneration, including autophagic disruption, organellar dysfunction, aging, and other cell stresses. In neurodegenerative disorders (e.g., Alzheimer's disease, Parkinson's disease, etc.), EVs contribute to the spread of pathological proteins like amyloid β, tau, ɑ-synuclein, prions, and TDP-43, exacerbating neurodegeneration and accelerating disease progression. Despite evidence for both neuropathological and neuroprotective effects of EVs, the mechanistic switch between their physiological and pathological functions remains elusive, warranting further research into their involvement in neurodegenerative disease. Moreover, owing to their innate ability to traverse the blood-brain barrier and their ubiquitous nature, EVs emerge as promising candidates for novel diagnostic and therapeutic strategies. The review uniquely positions itself at the intersection of EV cell biology, neurophysiology, and neuropathology, offering insights into the diverse biological roles of EVs in health and disease.
Collapse
Affiliation(s)
- Stephanie Tam
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Darcy Wear
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Christopher D Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Wai Haung Yu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Farahzadi R, Fathi E, Vandghanooni S, Valipour B. Hydrogel encapsulation of mesenchymal stem cells-derived extracellular vesicles as a novel therapeutic approach in cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189177. [PMID: 39218403 DOI: 10.1016/j.bbcan.2024.189177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Cell therapy has emerged as one of the most promising approaches to treating disease in recent decades. The application of stem cells in anti-tumor therapy is determined by their varying capacity for proliferation, migration, and differentiation. These capacities are derived from different sources. The use of stem cell carriers in cancer treatment is justified by the following three reasons: (I) shield therapeutic agents from swift biological deterioration; (II) reduce systemic side effects; and (III) increase local therapeutic levels since stem cells have an innate ability to target tumors. The quantity of stem cells confined to the tumor microenvironment determines this system's anti-tumor activity. Nevertheless, there are limitations to the use of different types of stem cells. When immune cells are used in cell therapy, it may lead to cytokine storms and improper reactions to self-antigens. Furthermore, the use of stem cells may result in cancer. Additionally, after an intravenous injection, cells could not migrate to the injury location. Exosomes derived from different cells were thus proposed as possible therapeutic options. Exosomes are becoming more and more well-liked because of their small size, biocompatibility, and simplicity in storage and separation. A number of investigations have shown that adding various medications and microRNAs to exosomes may enhance their therapeutic effectiveness. Thus, it is essential to evaluate studies looking into the therapeutic effectiveness of encapsulated exosomes. In this review, we looked at studies on encapsulated exosomes' use in regenerative medicine and the treatment of cancer. The results imply that the therapeutic potential increases when encapsulated exosomes are used rather than intact exosomes. Therefore, in order to optimize the effectiveness of the treatment, it is advised to implement this technique in accordance with the kind of therapy.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Valipour
- Department of Basic Sciences and Health, Sarab Faculty of Medical Sciences, Sarab, East Azerbaijan, Iran.
| |
Collapse
|
25
|
Mehta K, Yentsch H, Lee J, Yook Y, Lee KY, Gao TT, Tsai NP, Zhang K. Phosphatidylinositol-3-phosphate mediates Arc capsid secretion through the multivesicular body pathway. Proc Natl Acad Sci U S A 2024; 121:e2322422121. [PMID: 39178227 PMCID: PMC11363301 DOI: 10.1073/pnas.2322422121] [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: 01/03/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is an immediate early gene that plays a vital role in learning and memory. Arc protein has structural and functional properties similar to viral Group-specific antigen (Gag) protein and mediates the intercellular RNA transfer through virus-like capsids. However, the regulators and secretion pathway through which Arc capsids maneuver cargos are unclear. Here, we identified that phosphatidylinositol-3-phosphate (PI3P) mediates Arc capsid assembly and secretion through the endosomal-multivesicular body (MVB) pathway. Indeed, reconstituted Arc protein preferably binds to PI3P. In HEK293T cells, Arc forms puncta that colocalize with FYVE, an endosomal PI3P marker, as well as Rab5 and CD63, early endosomal and MVB markers, respectively. Superresolution imaging resolves Arc accumulates within the intraluminal vesicles of MVB. CRISPR double knockout of RalA and RalB, crucial GTPases for MVB biogenesis and exocytosis, severely reduces the Arc-mediated RNA transfer efficiency. RalA/B double knockdown in cultured rat cortical neurons increases the percentage of mature dendritic spines. Intake of extracellular vesicles purified from Arc-expressing wild-type, but not RalA/B double knockdown, cells in mouse cortical neurons reduces their surface GlutA1 levels. These results suggest that unlike the HIV Gag, whose membrane targeting requires interaction with plasma-membrane-specific phosphatidyl inositol (4,5) bisphosphate (PI(4,5)P2), the assembly of Arc capsids is mediated by PI3P at endocytic membranes. Understanding Arc's secretion pathway helps gain insights into its role in intercellular cargo transfer and highlights the commonality and distinction of trafficking mechanisms between structurally resembled capsid proteins.
Collapse
Affiliation(s)
- Kritika Mehta
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- NSF Science and Technology Center for Quantitative Cell Biology (STC-QCB) Center, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Henry Yentsch
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Jungbin Lee
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Yeeun Yook
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Tianyu Terry Gao
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Kai Zhang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- NSF Science and Technology Center for Quantitative Cell Biology (STC-QCB) Center, University of Illinois Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, IL61801
- Neuroscience Program, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL61801
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL61801
| |
Collapse
|
26
|
Liu Y, Sun L, Li Y, Holmes C. Mesenchymal stromal/stem cell tissue source and in vitro expansion impact extracellular vesicle protein and miRNA compositions as well as angiogenic and immunomodulatory capacities. J Extracell Vesicles 2024; 13:e12472. [PMID: 39092563 PMCID: PMC11294870 DOI: 10.1002/jev2.12472] [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: 12/21/2023] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Recently, therapies utilizing extracellular vesicles (EVs) derived from mesenchymal stromal/stem cells (MSCs) have begun to show promise in clinical trials. However, EV therapeutic potential varies with MSC tissue source and in vitro expansion through passaging. To find the optimal MSC source for clinically translatable EV-derived therapies, this study aims to compare the angiogenic and immunomodulatory potentials and the protein and miRNA cargo compositions of EVs isolated from the two most common clinical sources of adult MSCs, bone marrow and adipose tissue, across different passage numbers. Primary bone marrow-derived MSCs (BMSCs) and adipose-derived MSCs (ASCs) were isolated from adult female Lewis rats and expanded in vitro to the indicated passage numbers (P2, P4, and P8). EVs were isolated from the culture medium of P2, P4, and P8 BMSCs and ASCs and characterized for EV size, number, surface markers, protein content, and morphology. EVs isolated from different tissue sources showed different EV yields per cell, EV sizes, and protein yield per EV. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of proteomics data and miRNA seq data identified key proteins and pathways associated with differences between BMSC-EVs and ASC-EVs, as well as differences due to passage number. In vitro tube formation assays employing human umbilical vein endothelial cells suggested that both tissue source and passage number had significant effects on the angiogenic capacity of EVs. With or without lipopolysaccharide (LPS) stimulation, EVs more significantly impacted expression of M2-macrophage genes (IL-10, Arg1, TGFβ) than M1-macrophage genes (IL-6, NOS2, TNFα). By correlating the proteomics analyses with the miRNA seq analysis and differences observed in our in vitro immunomodulatory, angiogenic, and proliferation assays, this study highlights the trade-offs that may be necessary in selecting the optimal MSC source for development of clinical EV therapies.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Chemical & Biomedical Engineering, Florida A&M University‐Florida State University College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Li Sun
- Department of Chemical & Biomedical Engineering, Florida A&M University‐Florida State University College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- Department of Biomedical Sciences, College of MedicineFlorida State UniversityTallahasseeFloridaUSA
| | - Yan Li
- Department of Chemical & Biomedical Engineering, Florida A&M University‐Florida State University College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Christina Holmes
- Department of Chemical & Biomedical Engineering, Florida A&M University‐Florida State University College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| |
Collapse
|
27
|
Takakura Y, Hanayama R, Akiyoshi K, Futaki S, Hida K, Ichiki T, Ishii-Watabe A, Kuroda M, Maki K, Miura Y, Okada Y, Seo N, Takeuchi T, Yamaguchi T, Yoshioka Y. Quality and Safety Considerations for Therapeutic Products Based on Extracellular Vesicles. Pharm Res 2024; 41:1573-1594. [PMID: 39112776 PMCID: PMC11362369 DOI: 10.1007/s11095-024-03757-4] [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: 04/30/2024] [Accepted: 07/28/2024] [Indexed: 08/30/2024]
Abstract
Extracellular vesicles (EVs) serve as an intrinsic system for delivering functional molecules within our body, playing significant roles in diverse physiological phenomena and diseases. Both native and engineered EVs are currently the subject of extensive research as promising therapeutics and drug delivery systems, primarily due to their remarkable attributes, such as targeting capabilities, biocompatibility, and low immunogenicity and mutagenicity. Nevertheless, their clinical application is still a long way off owing to multiple limitations. In this context, the Science Board of the Pharmaceuticals and Medical Devices Agency (PMDA) of Japan has conducted a comprehensive assessment to identify the current issues related to the quality and safety of EV-based therapeutic products. Furthermore, we have presented several examples of the state-of-the-art methodologies employed in EV manufacturing, along with guidelines for critical processes, such as production, purification, characterization, quality evaluation and control, safety assessment, and clinical development and evaluation of EV-based therapeutics. These endeavors aim to facilitate the clinical application of EVs and pave the way for their transformative impact in healthcare.
Collapse
Affiliation(s)
- Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
| | - Rikinari Hanayama
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan.
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Kyoto, Japan
| | - Kyoko Hida
- Vascular Biology and Molecular Biology, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Takanori Ichiki
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Bunkyō, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kawasaki, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Shinjuku, Japan
| | - Kazushige Maki
- Pharmaceuticals and Medical Devices Agency, Chiyoda-ku, Japan
| | - Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Yoshiaki Okada
- Department of Transfusion Medicine and Cell Transplantation, Saitama Medical University Hospital, Kawagoe, Japan
| | - Naohiro Seo
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyō, Japan
| | - Toshihide Takeuchi
- Life Science Research Institute, Kindai University, Higashi-osaka, Japan
| | | | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Shinjuku, Japan
| |
Collapse
|
28
|
van der Koog L, Boerrigter MJ, Gorter IC, Gosens R, Nagelkerke A. Lung fibroblast-derived extracellular vesicles and soluble factors alleviate elastase-induced lung injury. Eur J Pharmacol 2024; 974:176612. [PMID: 38677537 DOI: 10.1016/j.ejphar.2024.176612] [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: 11/17/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
One of the main pathological features of chronic obstructive pulmonary disease (COPD) is the loss of functional alveolar tissue as a consequence of impaired regenerative capacities (emphysema). Recent research suggests that the secretome from mesenchymal cells, particularly extracellular vesicles (EVs), may possess regenerative properties beneficial for lung repair. However, the regenerative potential of the soluble factors (SFs) within the secretome remains largely unexplored in COPD. To this extent, we purified EVs and SFs secreted by lung fibroblasts to generate EV-enriched and SF-enriched fractions, and evaluated their effects on elastase-induced lung injury in both precision-cut lung slices (PCLS) and a mouse model. EV- and SF-enriched fractions were concentrated and purified from the conditioned medium of cultured MRC-5 lung fibroblasts using a combination of ultrafiltration and size exclusion chromatography, and were subsequently characterized according to the MISEV guidelines. Treatment with EV- or SF-enriched concentrates prevented and improved elastase-induced emphysema in PCLS, leading to reduced lung injury and upregulated markers of alveolar epithelial cells (aquaporin 5 and surfactant protein C), indicating potential parenchymal regeneration. Accordingly, prophylactic intratracheal treatment with lung fibroblast-derived EV- and SF-enriched concentrates in vivo attenuated elastase-induced lung tissue destruction, improved lung function, and enhanced gene expression of alveolar epithelial cell markers. Here, alveolar repair not only serves the purpose of facilitating gas exchange, but also by reinstating the essential parenchymal tethering required for optimal airway mechanics. In conclusion, this study highlights the therapeutic potential of both lung fibroblast-derived EV- and SF-enriched concentrates for the treatment of lung injury and emphysema.
Collapse
Affiliation(s)
- Luke van der Koog
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; GRIAC, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | - Myrthe J Boerrigter
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; GRIAC, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; GRIAC, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | - Anika Nagelkerke
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
29
|
Shi X, Hu X, Jiang N, Mao J. Regenerative endodontic therapy: From laboratory bench to clinical practice. J Adv Res 2024:S2090-1232(24)00267-4. [PMID: 38969092 DOI: 10.1016/j.jare.2024.07.001] [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: 04/16/2024] [Revised: 06/16/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Maintaining the vitality and functionality of dental pulp is paramount for tooth integrity, longevity, and homeostasis. Aiming to treat irreversible pulpitis and necrosis, there has been a paradigm shift from conventional root canal treatment towards regenerative endodontic therapy. AIM OF REVIEW This extensive and multipart review presents crucial laboratory and practical issues related to pulp-dentin complex regeneration aimed towards advancing clinical translation of regenerative endodontic therapy and enhancing human life quality. KEY SCIENTIFIC CONCEPTS OF REVIEW In this multipart review paper, we first present a panorama of emerging potential tissue engineering strategies for pulp-dentin complex regeneration from cell transplantation and cell homing perspectives, emphasizing the critical regenerative components of stem cells, biomaterials, and conducive microenvironments. Then, this review provides details about current clinically practiced pulp regenerative/reparative approaches, including direct pulp capping and root revascularization, with a specific focus on the remaining hurdles and bright prospects in developing such therapies. Next, special attention was devoted to discussing the innovative biomimetic perspectives opened in establishing functional tissues by employing exosomes and cell aggregates, which will benefit the clinical translation of dental pulp engineering protocols. Finally, we summarize careful consideration that should be given to basic research and clinical applications of regenerative endodontics. In particular, this review article highlights significant challenges associated with residual infection and inflammation and identifies future insightful directions in creating antibacterial and immunomodulatory microenvironments so that clinicians and researchers can comprehensively understand crucial clinical aspects of regenerative endodontic procedures.
Collapse
Affiliation(s)
- Xin Shi
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiaohan Hu
- Outpatient Department Office, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Nan Jiang
- Central Laboratory, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Jing Mao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| |
Collapse
|
30
|
Li J, Liu Y, Zhang R, Yang Q, Xiong W, He Y, Ye Q. Insights into the role of mesenchymal stem cells in cutaneous medical aesthetics: from basics to clinics. Stem Cell Res Ther 2024; 15:169. [PMID: 38886773 PMCID: PMC11184751 DOI: 10.1186/s13287-024-03774-5] [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: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
With the development of the economy and the increasing prevalence of skin problems, cutaneous medical aesthetics are gaining more and more attention. Skin disorders like poor wound healing, aging, and pigmentation have an impact not only on appearance but also on patients with physical and psychological issues, and even impose a significant financial burden on families and society. However, due to the complexities of its occurrence, present treatment options cannot produce optimal outcomes, indicating a dire need for new and effective treatments. Mesenchymal stem cells (MSCs) and their secretomics treatment is a new regenerative medicine therapy that promotes and regulates endogenous stem cell populations and/or replenishes cell pools to achieve tissue homeostasis and regeneration. It has demonstrated remarkable advantages in several skin-related in vivo and in vitro investigations, aiding in the improvement of skin conditions and the promotion of skin aesthetics. As a result, this review gives a complete description of recent scientific breakthroughs in MSCs for skin aesthetics and the limitations of their clinical applications, aiming to provide new ideas for future research and clinical transformation.
Collapse
Affiliation(s)
- Junyi Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Zhang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianyu Yang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
31
|
Shimizu Y, Ntege EH, Inoue Y, Matsuura N, Sunami H, Sowa Y. Optimizing mesenchymal stem cell extracellular vesicles for chronic wound healing: Bioengineering, standardization, and safety. Regen Ther 2024; 26:260-274. [PMID: 38978963 PMCID: PMC11228664 DOI: 10.1016/j.reth.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Chronic wounds represent a significant global burden, afflicting millions with debilitating complications. Despite standard care, impaired healing persists due to factors like persistent inflammation and impaired tissue regeneration. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) offer an innovative regenerative medicine approach, delivering stem cell-derived therapeutic cargo in engineered nanoscale delivery systems. This review examines pioneering bioengineering strategies to engineer MSC-EVs into precision nanotherapeutics for chronic wounds. Emerging technologies like CRISPR gene editing, microfluidic manufacturing, and biomimetic delivery systems are highlighted for their potential to enhance MSC-EV targeting, optimize therapeutic cargo enrichment, and ensure consistent clinical-grade production. However, key hurdles remain, including batch variability, rigorous safety assessment for potential tumorigenicity, immunogenicity, and biodistribution profiling. Crucially, collaborative frameworks harmonizing regulatory science with bioengineering and patient advocacy hold the key to expediting global clinical translation. By overcoming these challenges, engineered MSC-EVs could catalyze a new era of off-the-shelf regenerative therapies, restoring hope and healing for millions afflicted by non-healing wounds.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| |
Collapse
|
32
|
Wang S, Jia Z, Dai M, Feng X, Tang C, Liu L, Cao L. Advances in natural and synthetic macromolecules with stem cells and extracellular vesicles for orthopedic disease treatment. Int J Biol Macromol 2024; 268:131874. [PMID: 38692547 DOI: 10.1016/j.ijbiomac.2024.131874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Serious orthopedic disorders resulting from myriad diseases and impairments continue to pose a considerable challenge to contemporary clinical care. Owing to its limited regenerative capacity, achieving complete bone tissue regeneration and complete functional restoration has proven challenging with existing treatments. By virtue of cellular regenerative and paracrine pathways, stem cells are extensively utilized in the restoration and regeneration of bone tissue; however, low survival and retention after transplantation severely limit their therapeutic effect. Meanwhile, biomolecule materials provide a delivery platform that improves stem cell survival, increases retention, and enhances therapeutic efficacy. In this review, we present the basic concepts of stem cells and extracellular vesicles from different sources, emphasizing the importance of using appropriate expansion methods and modification strategies. We then review different types of biomolecule materials, focusing on their design strategies. Moreover, we summarize several forms of biomaterial preparation and application strategies as well as current research on biomacromolecule materials loaded with stem cells and extracellular vesicles. Finally, we present the challenges currently impeding their clinical application for the treatment of orthopedic diseases. The article aims to provide researchers with new insights for subsequent investigations.
Collapse
Affiliation(s)
- Supeng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China; Ningxia Medical University, Ningxia 750004, China
| | - Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Xujun Feng
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Lingling Cao
- Jiujiang City Key Laboratory of Cell Therapy, The First Hospital of Jiujiang City, Jiujiang 332000, China.
| |
Collapse
|
33
|
Anvari Y, Afrashteh A, Pourkaveh S, Salek SB, Al-Numan L, Khademnezhad S. Emerging role of mesenchymal stem cell-derived extracellular vesicles in periodontal regeneration. J Taibah Univ Med Sci 2024; 19:390-402. [PMID: 38380419 PMCID: PMC10876597 DOI: 10.1016/j.jtumed.2024.01.006] [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: 10/25/2023] [Revised: 12/17/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
Periodontitis is a prevalent oral ailment that harms both hard and soft tissues of the periodontium, leading to loosening and eventual removal of the teeth. Current clinical treatments have limitations in achieving complete periodontal tissue regeneration. Mesenchymal stem cells (MSCs) have garnered attention due to their unique characteristics and potential as a promising new therapy for periodontitis. Research suggests that the role of MSCs in regenerative medicine primarily occurs through the paracrine pathway, involving the emission of particles encased by lipids called extracellular vesicles (EVs) abundant in bioactive compounds. These EVs play a vital function in controlling the activities of periodontal tissues and immune system cells, and by influencing the immediate surrounding, thus fostering the healing of periodontal damage and renewal of tissues. EVs obtained from MSCs (MSC-EVs), in the form of a cell-free treatment, offer advantages in terms of stability, reduced immune rejection, and ethical considerations, elevating their potential as a hopeful choice for broad clinical applications. This concise overview highlights the mechanisms of MSC-EVs and the possibilities they hold in clinical application for periodontal regeneration.
Collapse
Affiliation(s)
- Yaldasadat Anvari
- Department of Dentistry, School of Dentistry, Near East University, Nicosia, Cyprus
| | - Ahmad Afrashteh
- Department of Periodontics, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Pourkaveh
- Department of Periodontics, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira B. Salek
- Department of Periodontics, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Lelaw Al-Numan
- Department of Dentistry, School of Dentistry, Near East University, Nicosia, Cyprus
| | - Sahar Khademnezhad
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Zhang S, Liang Y, Ji P, Zheng R, Lu F, Hou G, Yang G, Yuan L. Truncated PD1 Engineered Gas-Producing Extracellular Vesicles for Ultrasound Imaging and Subsequent Degradation of PDL1 in Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305891. [PMID: 38263860 PMCID: PMC10966526 DOI: 10.1002/advs.202305891] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/05/2024] [Indexed: 01/25/2024]
Abstract
PDL1 blockade therapy holds great promise in cancer immunotherapy. Ultrasound imaging of PDL1 expression in the tumor is of great importance in predicting the therapeutic efficacy. As a proof-of-concept study, a novel ultrasound contrast agent has been innovated here to image and block PDL1 in the tumor tissue. Briefly, extracellular vesicles (EVs) are engineered to display truncated PD1 (tPD1) on the surface to bind PDL1 with high affinity by fusion to EV-abundant transmembrane protein PTGFRN. The engineered EVs are then encapsulated with Ca(HCO3)2 via electroporation and designated as Gp-EVtPD1, which would recognize PDL1 highly expressed cells and produce gas in the endosomes and lysosomes. On the one hand, the echogenic signal intensity correlates well with the PDL1 expression and immune response inhibition in the tumor. On the other hand, during the trajectory of Gp-EVtPD1 in the recipient cells, tPD1 on the EV binds PDL1 and triggers the PDL1 endocytosis and degradation in endosomes/lysosomes in a sequential manner, and thus boosts the anti-tumor immunity of cytotoxic T cells. In summary, Gp-EVtPD1 serves as a novel ultrasound contrast agent and blocker of PDL1, which might be of great advantage in imaging PDL1 expression and conquering immune checkpoint blocker resistance.
Collapse
Affiliation(s)
- Siyan Zhang
- Department of Ultrasound DiagnosticsTangdu HospitalFourth Military Medical UniversityXinsi Road No. 569thXi'an710038P. R. China
| | - Yuan Liang
- Department of Ultrasound DiagnosticsTangdu HospitalFourth Military Medical UniversityXinsi Road No. 569thXi'an710038P. R. China
| | - Panpan Ji
- Department of Digestive SurgeryXijing HospitalFourth Military Medical UniversityShaanxi710032P. R. China
| | - Rui Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular BiologyFourth Military Medical UniversityChanglexi Road No. 169thXi'an710032P. R. China
| | - Fan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular BiologyFourth Military Medical UniversityChanglexi Road No. 169thXi'an710032P. R. China
| | - Guangdong Hou
- Department of UrologyXijing HospitalFourth Military Medical UniversityXi'an710032P. R. China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular BiologyFourth Military Medical UniversityChanglexi Road No. 169thXi'an710032P. R. China
| | - Lijun Yuan
- Department of Ultrasound DiagnosticsTangdu HospitalFourth Military Medical UniversityXinsi Road No. 569thXi'an710038P. R. China
| |
Collapse
|
35
|
Sun F, Sun Y, Wang X, Zhu J, Chen S, Yu Y, Zhu M, Xu W, Qian H. Engineered mesenchymal stem cell-derived small extracellular vesicles for diabetic retinopathy therapy through HIF-1α/EZH2/PGC-1α pathway. Bioact Mater 2024; 33:444-459. [PMID: 38076648 PMCID: PMC10697990 DOI: 10.1016/j.bioactmat.2023.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/17/2023] [Accepted: 11/15/2023] [Indexed: 09/04/2024] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness worldwide with limited treatment options. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) hold promise as a cell-free therapy for retinal diseases. In this study, we present evidence that the intravitreal injection of MSC-sEVs improved retinal function and alleviated retinal apoptosis, inflammation, and angiogenesis in both db/db mice and streptozotocin-induced diabetic rats. Mechanistically, hyperglycemia-induced activation of hypoxia-inducible factor-1α (HIF-1α) inhibited the tripartite motif 21 (TRIM21)-mediated ubiquitination and degradation of enhancer of zeste homologue 2 (EZH2), ultimately resulting in the downregulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) through EZH2-induced methylation modification. The presence of miR-5068 and miR-10228 in MSC-sEVs targeted the HIF-1α/EZH2/PGC-1α pathway. The blockade of miR-5068 and miR-10228 abolished the retinal therapeutic effects of MSC-sEVs. Additionally, we engineered MSC-sEVs with elevated levels of miR-5068 and miR-10228 to enhance retinal repair efficiency. Together, our findings provide novel insights into the mechanism underlying DR progress and highlight the potential of MSC-sEVs, especially engineered MSC-sEVs, as a therapeutic option for DR.
Collapse
Affiliation(s)
- Fengtian Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China
| | - Yuntong Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China
| | - Xiaoling Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- Department of Laboratory Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361006, Fujian, China
| | - Junyan Zhu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Shenyuan Chen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yifan Yu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Mengyao Zhu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology & Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, 200040, China
| |
Collapse
|
36
|
Ju Y, Yang P, Liu X, Qiao Z, Shen N, Lei L, Fang B. Microenvironment Remodeling Self-Healing Hydrogel for Promoting Flap Survival. Biomater Res 2024; 28:0001. [PMID: 38390027 PMCID: PMC10882600 DOI: 10.34133/bmr.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/13/2023] [Indexed: 02/24/2024] Open
Abstract
Random flap grafting is a routine procedure used in plastic and reconstructive surgery to repair and reconstruct large tissue defects. Flap necrosis is primarily caused by ischemia-reperfusion injury and inadequate blood supply to the distal flap. Ischemia-reperfusion injury leads to the production of excessive reactive oxygen species, creating a pathological microenvironment that impairs cellular function and angiogenesis. In this study, we developed a microenvironment remodeling self-healing hydrogel [laminarin-chitosan-based hydrogel-loaded extracellular vesicles and ceria nanozymes (LCH@EVs&CNZs)] to improve the flap microenvironment and synergistically promote flap regeneration and survival. The natural self-healing hydrogel (LCH) was created by the oxidation laminarin and carboxymethylated chitosan via a Schiff base reaction. We loaded this hydrogel with CNZs and EVs. CNZs are a class of nanomaterials with enzymatic activity known for their strong scavenging capacity for reactive oxygen species, thus alleviating oxidative stress. EVs are cell-secreted vesicular structures containing thousands of bioactive substances that can promote cell proliferation, migration, differentiation, and angiogenesis. The constructed LCH@EVs&CNZs demonstrated a robust capacity for scavenging excess reactive oxygen species, thereby conferring cellular protection in oxidative stress environments. Moreover, these constructs notably enhance cell migration and angiogenesis. Our results demonstrate that LCH@EVs&CNZs effectively remodel the pathological skin flap microenvironment and marked improve flap survival. This approach introduces a new therapeutic strategy combining microenvironmental remodeling with EV therapy, which holds promise for promoting flap survival.
Collapse
Affiliation(s)
- Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital,
Central South University, Changsha, Hunan 410011, China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital,
Central South University, Changsha, Hunan 410011, China
| | - Xiangjun Liu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital,
Central South University, Changsha, Hunan 410011, China
| | - Zhihua Qiao
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital,
Central South University, Changsha, Hunan 410011, China
| | - Naisi Shen
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital,
Central South University, Changsha, Hunan 410011, China
| | - Lanjie Lei
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou, Zhejiang 310015, China
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital,
Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
37
|
Berry D, Ene J, Nathani A, Singh M, Li Y, Zeng C. Effects of Physical Cues on Stem Cell-Derived Extracellular Vesicles toward Neuropathy Applications. Biomedicines 2024; 12:489. [PMID: 38540102 PMCID: PMC10968089 DOI: 10.3390/biomedicines12030489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 11/28/2024] Open
Abstract
The peripheral nervous system undergoes sufficient stress when affected by diabetic conditions, chemotherapeutic drugs, and personal injury. Consequently, peripheral neuropathy arises as the most common complication, leading to debilitating symptoms that significantly alter the quality and way of life. The resulting chronic pain requires a treatment approach that does not simply mask the accompanying symptoms but provides the necessary external environment and neurotrophic factors that will effectively facilitate nerve regeneration. Under normal conditions, the peripheral nervous system self-regenerates very slowly. The rate of progression is further hindered by the development of fibrosis and scar tissue formation, which does not allow sufficient neurite outgrowth to the target site. By incorporating scaffolding supplemented with secretome derived from human mesenchymal stem cells, it is hypothesized that neurotrophic factors and cellular signaling can facilitate the optimal microenvironment for nerve reinnervation. However, conventional methods of secretory vesicle production are low yield, thus requiring improved methods to enhance paracrine secretions. This report highlights the state-of-the-art methods of neuropathy treatment as well as methods to optimize the clinical application of stem cells and derived secretory vesicles for nerve regeneration.
Collapse
Affiliation(s)
- Danyale Berry
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida Agricultural and Mechanical University, Tallahassee, FL 32310, USA;
- High Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 23210, USA
| | - Justice Ene
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida Agricultural and Mechanical University, Tallahassee, FL 32307, USA; (A.N.); (M.S.)
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida Agricultural and Mechanical University, Tallahassee, FL 32307, USA; (A.N.); (M.S.)
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
| | - Changchun Zeng
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida Agricultural and Mechanical University, Tallahassee, FL 32310, USA;
- High Performance Materials Institute, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 23210, USA
| |
Collapse
|
38
|
Gao H, Liu S, Qin S, Yang J, Yue T, Ye B, Tang Y, Feng J, Hou J, Danzeng D. Injectable hydrogel-based combination therapy for myocardial infarction: a systematic review and Meta-analysis of preclinical trials. BMC Cardiovasc Disord 2024; 24:119. [PMID: 38383333 PMCID: PMC10882925 DOI: 10.1186/s12872-024-03742-0] [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: 09/30/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION This study evaluates the effectiveness of a combined regimen involving injectable hydrogels for the treatment of experimental myocardial infarction. PATIENT CONCERNS Myocardial infarction is an acute illness that negatively affects quality of life and increases mortality rates. Experimental models of myocardial infarction can aid in disease research by allowing for the development of therapies that effectively manage disease progression and promote tissue repair. DIAGNOSIS Experimental animal models of myocardial infarction were established using the ligation method on the anterior descending branch of the left coronary artery (LAD). INTERVENTIONS The efficacy of intracardiac injection of hydrogels, combined with cells, drugs, cytokines, extracellular vesicles, or nucleic acid therapies, was evaluated to assess the functional and morphological improvements in the post-infarction heart achieved through the combined hydrogel regimen. OUTCOMES A literature review was conducted using PubMed, Web of Science, Scopus, and Cochrane databases. A total of 83 papers, including studies on 1332 experimental animals (rats, mice, rabbits, sheep, and pigs), were included in the meta-analysis based on the inclusion and exclusion criteria. The overall effect size observed in the group receiving combined hydrogel therapy, compared to the group receiving hydrogel treatment alone, resulted in an ejection fraction (EF) improvement of 8.87% [95% confidence interval (CI): 7.53, 10.21] and a fractional shortening (FS) improvement of 6.31% [95% CI: 5.94, 6.67] in rat models, while in mice models, the improvements were 16.45% [95% CI: 11.29, 21.61] for EF and 5.68% [95% CI: 5.15, 6.22] for FS. The most significant improvements in EF (rats: MD = 9.63% [95% CI: 4.02, 15.23]; mice: MD = 23.93% [95% CI: 17.52, 30.84]) and FS (rats: MD = 8.55% [95% CI: 2.54, 14.56]; mice: MD = 5.68% [95% CI: 5.15, 6.22]) were observed when extracellular vesicle therapy was used. Although there have been significant results in large animal experiments, the number of studies conducted in this area is limited. CONCLUSION The present study demonstrates that combining hydrogel with other therapies effectively improves heart function and morphology. Further preclinical research using large animal models is necessary for additional study and validation.
Collapse
Affiliation(s)
- Han Gao
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Song Liu
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Shanshan Qin
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Bengui Ye
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yue Tang
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Feng
- School of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Cardiology, Chengdu Third People's Hospital, Chengdu, Sichuan, China.
| | - Dunzhu Danzeng
- School of Medicine, Tibet University, Lhasa, Tibet, China.
| |
Collapse
|
39
|
He X, Guo C, Wang Y, Ma S, Liu X, Wei Y, Xu H, Liang Z, Hu Y, Zhao L, Lian X, Huang D. Enhancing osseointegration of titanium implants through MC3T3-E1 protein-gelatin polyelectrolyte multilayers. J Biomed Mater Res B Appl Biomater 2024; 112:e35373. [PMID: 38359169 DOI: 10.1002/jbm.b.35373] [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: 06/08/2023] [Revised: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 02/17/2024]
Abstract
Titanium and its alloys have found extensive use in the biomedical field, however, implant loosening due to weak osseointegration remains a concern. Improved surface morphology and chemical composition can enhance the osseointegration of the implant. Bioactive molecules have been utilized to modify the surface of the titanium-based material to achieve rapid and efficient osseointegration between the implant and bone tissues. In this study, the bioactive substance MC3T3-E1 protein-gelatin polyelectrolyte multilayers were constructed on the surface of the titanium implants by means of layer-by-layer self-assembly to enhance the strength of the bond between the bone tissue and the implant. The findings of the study indicate that the layer-by-layer self-assembly technique can enhance surface roughness and hydrophilicity to a considerable extent. Compared to pure titanium, the hydrophilicity of TiOH LBL was significantly increased with a water contact angle of 75.0 ± $$ \pm $$ 2.4°. The modified titanium implant exhibits superior biocompatibility and wound healing ability upon co-culture with cells. MC3T3-E1 cells were co-cultured with TiOH LBL for 1, 3, and 5 days and their viability was higher than 85%. In addition, the wound healing results demonstrate that TiOH LBL exhibited the highest migratory ability (243 ± 10 μm). Furthermore, after 7 days of osteogenic induction, the modified titanium implant significantly promotes osteoblast differentiation.
Collapse
Affiliation(s)
- Xuhong He
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
| | - Chaiqiong Guo
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
| | - Yuhui Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
| | - Shilong Ma
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
| | - Xuanyu Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| | - Haofeng Xu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
| | - Ziwei Liang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| | - Yinchun Hu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| |
Collapse
|
40
|
Marquez CA, Oh CI, Ahn G, Shin WR, Kim YH, Ahn JY. Synergistic vesicle-vector systems for targeted delivery. J Nanobiotechnology 2024; 22:6. [PMID: 38167116 PMCID: PMC10763086 DOI: 10.1186/s12951-023-02275-6] [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: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
With the immense progress in drug delivery systems (DDS) and the rise of nanotechnology, challenges such as target specificity remain. The vesicle-vector system (VVS) is a delivery system that uses lipid-based vesicles as vectors for a targeted drug delivery. When modified with target-probing materials, these vesicles become powerful vectors for drug delivery with high target specificity. In this review, we discuss three general types of VVS based on different modification strategies: (1) vesicle-probes; (2) vesicle-vesicles; and (3) genetically engineered vesicles. The synthesis of each VVS type and their corresponding properties that are advantageous for targeted drug delivery, are also highlighted. The applications, challenges, and limitations of VVS are briefly examined. Finally, we share a number of insights and perspectives regarding the future of VVS as a targeted drug delivery system at the nanoscale.
Collapse
Affiliation(s)
- Christine Ardelle Marquez
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Cho-Im Oh
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Gna Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Woo-Ri Shin
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Department of Bioengineering, University of Pennsylvania, 210 S 33rd St, Philadelphia, PA, 19104, USA
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Ji-Young Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
41
|
Shi H, Yang Y, Xing H, Jia J, Xiong W, Guo S, Yang S. Exosomal non-coding RNAs: Emerging insights into therapeutic potential and mechanisms in bone healing. J Tissue Eng 2024; 15:20417314241286606. [PMID: 39371940 PMCID: PMC11456177 DOI: 10.1177/20417314241286606] [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: 08/11/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Exosomes are nano-sized extracellular vesicles (EVs) released by diverse types of cells, which affect the functions of targeted cells by transporting bioactive substances. As the main component of exosomes, non-coding RNA (ncRNA) is demonstrated to impact multiple pathways participating in bone healing. Herein, this review first introduces the biogenesis and secretion of exosomes, and elucidates the role of the main cargo in exosomes, ncRNAs, in mediating intercellular communication. Subsequently, the potential molecular mechanism of exosomes accelerating bone healing is elucidated from the following four aspects: macrophage polarization, vascularization, osteogenesis and osteoclastogenesis. Then, we systematically introduce construction strategies based on modified exosomes in bone regeneration field. Finally, the clinical trials of exosomes for bone healing and the challenges of exosome-based therapies in the biomedical field are briefly introduced, providing solid theoretical frameworks and optimization methods for the clinical application of exosomes in orthopedics.
Collapse
Affiliation(s)
- Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yang Yang
- Department of Rehabilitation, The First Hospital of China Medical University, Shenyang, China
| | - Hao Xing
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jialin Jia
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
42
|
Allahham N, Colic I, Rayner MLD, Gurnani P, Phillips JB, Rahim AA, Williams GR. Advanced Formulation Approaches for Emerging Therapeutic Technologies. Handb Exp Pharmacol 2024; 284:343-365. [PMID: 37733107 DOI: 10.1007/164_2023_695] [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: 09/22/2023]
Abstract
In addition to proteins, discussed in the Chapter "Advances in Vaccine Adjuvants: Nanomaterials and Small Molecules", there are a wide range of alternatives to small molecule active ingredients. Cells, extracellular vesicles, and nucleic acids in particular have attracted increasing research attention in recent years. There are now a number of products on the market based on these emerging technologies, the most famous of which are the mRNA-based vaccines against SARS-COV-2. These advanced therapeutic moieties are challenging to formulate however, and there remain significant challenges for their more widespread use. In this chapter, we consider the potential and bottlenecks for developing further medical products based on these systems. Cells, extracellular vesicles, and nucleic acids will be discussed in terms of their mechanism of action, the key requirements for translation, and how advanced formulation approaches can aid their future development. These points will be presented with selected examples from the literature, and with a focus on the formulations which have made the transition to clinical trials and clinical products.
Collapse
Affiliation(s)
- Nour Allahham
- UCL School of Pharmacy, University College London, London, UK
| | - Ines Colic
- UCL School of Pharmacy, University College London, London, UK
| | | | - Pratik Gurnani
- UCL School of Pharmacy, University College London, London, UK
| | | | - Ahad A Rahim
- UCL School of Pharmacy, University College London, London, UK
| | | |
Collapse
|
43
|
Bai J, Yu B, Li C, Cheng H, Guan Y, Ren Z, Zhang T, Song X, Jia Z, Su T, Tao B, Gao H, Yang B, Liang L, Xiong X, Zhou X, Yin L, Peng J, Shang A, Wang Y. Mesenchymal Stem Cell-Derived Mitochondria Enhance Extracellular Matrix-Derived Grafts for the Repair of Nerve Defect. Adv Healthc Mater 2024; 13:e2302128. [PMID: 37922434 PMCID: PMC11468562 DOI: 10.1002/adhm.202302128] [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: 07/06/2023] [Revised: 10/27/2023] [Indexed: 11/05/2023]
Abstract
Peripheral nerve injuries (PNI) can lead to mitochondrial dysfunction and energy depletion within the affected microenvironment. The objective is to investigate the potential of transplanting mitochondria to reshape the neural regeneration microenvironment. High-purity functional mitochondria with an intact structure are extracted from human umbilical cord-derived mesenchymal stem cells (hUCMSCs) using the Dounce homogenization combined with ultracentrifugation. Results show that when hUCMSC-derived mitochondria (hUCMSC-Mitos) are cocultured with Schwann cells (SCs), they promote the proliferation, migration, and respiratory capacity of SCs. Acellular nerve allografts (ANAs) have shown promise in nerve regeneration, however, their therapeutic effect is not satisfactory enough. The incorporation of hUCMSC-Mitos within ANAs has the potential to remodel the regenerative microenvironment. This approach demonstrates satisfactory outcomes in terms of tissue regeneration and functional recovery. Particularly, the use of metabolomics and bioenergetic profiling is used for the first time to analyze the energy metabolism microenvironment after PNI. This remodeling occurs through the enhancement of the tricarboxylic acid cycle and the regulation of associated metabolites, resulting in increased energy synthesis. Overall, the hUCMSC-Mito-loaded ANAs exhibit high functionality to promote nerve regeneration, providing a novel regenerative strategy based on improving energy metabolism for neural repair.
Collapse
Affiliation(s)
- Jun Bai
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Co‐innovation Center of NeuroregenerationNantong University NantongJiangsu Province226007P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Bingbing Yu
- School of Materials Science and EngineeringThe Key Laboratory of Advanced Materials of Ministry of EducationState Key Laboratory of New Ceramics and Fine ProcessingCenter for Flexible Electronics TechnologyTsinghua UniversityBeijing100084P. R. China
| | - Chaochao Li
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Haofeng Cheng
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
- School of MedicineNankai UniversityTianjin300071P. R. China
| | - Yanjun Guan
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Zhiqi Ren
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Tieyuan Zhang
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Xiangyu Song
- School of MedicineHebei North UniversityZhangjiakou075051P. R. China
| | - Zhibo Jia
- School of MedicineHebei North UniversityZhangjiakou075051P. R. China
| | - Tianqi Su
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Benzhang Tao
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Haihao Gao
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Boyao Yang
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Lijing Liang
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Xing Xiong
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Xingyu Zhou
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
- Graduate School of Chinese PLA General HospitalNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Lan Yin
- School of Materials Science and EngineeringThe Key Laboratory of Advanced Materials of Ministry of EducationState Key Laboratory of New Ceramics and Fine ProcessingCenter for Flexible Electronics TechnologyTsinghua UniversityBeijing100084P. R. China
| | - Jiang Peng
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Co‐innovation Center of NeuroregenerationNantong University NantongJiangsu Province226007P. R. China
| | - Aijia Shang
- Department of NeurosurgeryGeneral Hospital of Chinese People Liberty ArmyNo. 28 Fuxing RoadBeijing100853P. R. China
| | - Yu Wang
- Institute of OrthopedicsThe Fourth Medical Center of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma and War Injuries PLANo. 51 Fucheng RoadBeijing100048P. R. China
- Co‐innovation Center of NeuroregenerationNantong University NantongJiangsu Province226007P. R. China
| |
Collapse
|
44
|
Mehta K, Yentsch H, Lee J, Gao TT, Zhang K. Phosphatidylinositol 3-phosphate mediates Arc capsids secretion through the multivesicular body pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572392. [PMID: 38187623 PMCID: PMC10769229 DOI: 10.1101/2023.12.19.572392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is an immediate early gene that plays a vital role in learning and memory. The recent discovery that Arc mediates the inter-neuronal RNA transfer implies its role in regulating neuronal functions across long distances. Arc protein has structural and functional properties similar to viral Group-specific antigen (Gag). By assembling into high-order, virus-like capsids, Arc mediates the intercellular RNA transfer. However, the exact secretion pathway through which Arc capsids maneuver cargos is unclear. Here, we identified that Arc capsids assemble and secrete through the endosomal-multivesicular body (MVB) pathway. Arc's endosomal entry is likely mediated by phosphatidylinositol-3-phosphate (PI3P). Indeed, reconstituted Arc protein preferably binds to PI3P. In mammalian cells, Arc forms puncta that colocalizes with FYVE, an endosomal PI3P marker, and competitive binding to PI3P via prolonged FYVE expression reduces the average number of Arc puncta per cell. Overexpression of MTMR1, a PI3P phosphatase, significantly reduces Arc capsid secretion. Arc capsids secrete through the endosomal-MVB axis as extracellular vesicles. Live-cell imaging shows that fluorescently labeled Arc primarily colocalizes Rab5 and CD63, early endosomal and MVB markers, respectively. Superresolution imaging resolves Arc accumulates within the intraluminal vesicles of MVB. CRISPR double knockout of RalA and RalB, crucial GTPases for MVB biogenesis and exocytosis, severely reduces Arc-mediated RNA transfer efficiency. These results suggest that, unlike the Human Immunodeficiency Virus Gag, which assembles on and bud off from the plasma membrane, Arc capsids assemble at the endocytic membranes of the endosomal-MVB pathway mediated by PI3P. Understanding Arc's secretion pathway helps gain insights into its role in intercellular cargo transfer and highlights the commonality and distinction of trafficking mechanisms between structurally resembled capsid proteins.
Collapse
|
45
|
Zhang T, Wu Y, Li X, Zhang A, Liu H, Shi M, Zhang Z, Lu W, Guo Y, Tang X, Cui Q, Li Z. Small extracellular vesicles derived from tendon stem cells promote the healing of injured Achilles tendons by regulating miR-145-3p. Acta Biomater 2023; 172:280-296. [PMID: 37806377 DOI: 10.1016/j.actbio.2023.10.004] [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: 04/18/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
The therapeutic role of tendon stem cells (TSCs) in tendon-related injuries has been well documented. Small extracellular vesicles (sEVs) are being increasingly used as new biotherapeutic agents for various diseases. Therefore, the potential function of TSC-sEVs in tendon injury repair warrants further investigation. In this study, we explored the effects of TSC-sEVs on TSC proliferation, migration, and differentiation in vitro in an autocrine manner. We further used a novel exosomal topical treatment with TSC-sEVs loaded with gelatin methacryloyl (GelMA) hydrogel in vivo; we mixed sufficient amounts of TSC-sEVs with GelMA hydrogel to cover the damaged molded Achilles tendon tissue and then exposed them to UV irradiation for coagulation. GelMA loading ensured that TSC-sEVs were slowly released at the injury site over a long period, thereby achieving their full local therapeutic effects. Treatment with TSC-sEVs loaded with GelMA significantly improved the histological score of the regenerated tendon by increasing the tendon expression while inhibiting the formation of excessive ossification and improving the mechanical properties of the tissue. Moreover, miRNA sequencing in TSC-sEVs, TSCs, and TSCs receiving sEVs revealed that TSC-sEVs altered the miRNA expression profile of TSCs, with increased expression of miR-145-3p. In conclusion, our study demonstrates that TSC-sEVs can play a key role in treating tendon injuries and that loading them with GelMA can enhance their effect in vivo. Moreover, miR-145-3p has a major functional role in the effect of TSC-sEVs. This study offers new therapeutic ideas for the local treatment of Achilles tendon injuries using sEVs. STATEMENT OF SIGNIFICANCE: In this study, we demonstrated that TSC-sEVs play a key role in treating tendon injuries and that loading them with GelMA hydrogel can act as a fixation and slow release in vivo. Moreover, it identifies the major functional role of miR-145-3p in the effect of TSCs that were identified and validated by miRNA sequencing. Our study provides a basis for further research on GelMA slow-release assays that have potential clinical applications. It offers new therapeutic ideas for the local treatment of Achilles tendon injuries using TSC-sEVs.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of General Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Yang Wu
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China
| | - Xiangqi Li
- Department of General Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Aodan Zhang
- Department of General Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Hengchen Liu
- Department of General Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, PR China
| | - Manyu Shi
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China
| | - Zenan Zhang
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China
| | - Wenjun Lu
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China
| | - Yujun Guo
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China
| | - Xin Tang
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China
| | - Qingbo Cui
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China.
| | - Zhaozhu Li
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150028, PR China.
| |
Collapse
|
46
|
Min K, Sahu A, Jeon SH, Tae G. Emerging drug delivery systems with traditional routes - A roadmap to chronic inflammatory diseases. Adv Drug Deliv Rev 2023; 203:115119. [PMID: 37898338 DOI: 10.1016/j.addr.2023.115119] [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: 09/15/2022] [Revised: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Inflammation is prevalent and inevitable in daily life but can generally be accommodated by the immune systems. However, incapable self-healing and persistent inflammation can progress to chronic inflammation, leading to prevalent or fatal chronic diseases. This review comprehensively covers the topic of emerging drug delivery systems (DDSs) for the treatment of chronic inflammatory diseases (CIDs). First, we introduce the basic biology of the chronic inflammatory process and provide an overview of the main CIDs of the major organs. Next, up-to-date information on various DDSs and the associated strategies for ensuring targeted delivery and stimuli-responsiveness applied to CIDs are discussed extensively. The implementation of traditional routes of drug administration to maximize their therapeutic effects against CIDs is then summarized. Finally, perspectives on future DDSs against CIDs are presented.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Hajipur, 844102, India
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
47
|
Yang T, Zhao F, Zhao J, Geng J, Shao C, Liu J, Sheng F, Zhou L, Xu H, Jia R. Negatively charged bladder acellular matrix loaded with positively charged adipose-derived mesenchymal stem cell-derived small extracellular vesicles for bladder tissue engineering. J Control Release 2023; 364:718-733. [PMID: 37944669 DOI: 10.1016/j.jconrel.2023.10.048] [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: 05/14/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Adipose-derived mesenchymal stem cell-derived small extracellular vesicles (Ad-MSC-sEVs/AMEs) combined with scaffold materials are used in tissue-engineered bladders; however, the lack of retention leads to limited distribution of AMEs in the scaffold areas and low bioavailability of AMEs after bladder reconstruction. To improve retention of AMEs, we developed a novel strategy that modifies the surface charge of the bladder acellular matrix (BAM) via oxidative self-polymerization of dopamine-reducing graphene oxide (GO) and AMEs using ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD). We evaluated two BAM surface modification methods and evaluated the biocompatibility of materials and PPD and electrostatic adherence effects between PPD-modified AMEs and rGO-PDA/BAM in vivo and in vitro. Surface modification increased retention of AMEs, enhanced regeneration of bladder structures, and increased electrical conductivity of rGO-PDA/BAM, thereby improving bladder function recovery. RNA-sequencing revealed 543 miRNAs in human AMEs and 514 miRNAs in rat AMEs. A Venn diagram was used to show target genes of miRNA with the highest proportion predicted by the four databases; related biological processes and pathways were predicted by KEGG and GO analyses. We report a strategy for improving bioavailability of AMEs for bladder reconstruction and reveal that enriched miR-21-5p targets PIK3R1 and activates the PI3K/Akt pathway to promote cell proliferation and migration.
Collapse
Affiliation(s)
- Tianli Yang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jun Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jian Geng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Si Pai Lou 2, Nanjing 210096, China
| | - Cheng Shao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Fei Sheng
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| | - Hua Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Si Pai Lou 2, Nanjing 210096, China.
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| |
Collapse
|
48
|
Xu P, Kankala RK, Wang S, Chen A. Decellularized extracellular matrix-based composite scaffolds for tissue engineering and regenerative medicine. Regen Biomater 2023; 11:rbad107. [PMID: 38173774 PMCID: PMC10761212 DOI: 10.1093/rb/rbad107] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/17/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Despite the considerable advancements in fabricating polymeric-based scaffolds for tissue engineering, the clinical transformation of these scaffolds remained a big challenge because of the difficulty of simulating native organs/tissues' microenvironment. As a kind of natural tissue-derived biomaterials, decellularized extracellular matrix (dECM)-based scaffolds have gained attention due to their unique biomimetic properties, providing a specific microenvironment suitable for promoting cell proliferation, migration, attachment and regulating differentiation. The medical applications of dECM-based scaffolds have addressed critical challenges, including poor mechanical strength and insufficient stability. For promoting the reconstruction of damaged tissues or organs, different types of dECM-based composite platforms have been designed to mimic tissue microenvironment, including by integrating with natural polymer or/and syntenic polymer or adding bioactive factors. In this review, we summarized the research progress of dECM-based composite scaffolds in regenerative medicine, highlighting the critical challenges and future perspectives related to the medical application of these composite materials.
Collapse
Affiliation(s)
- Peiyao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Shibin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, PR China
| |
Collapse
|
49
|
Lee CS, Lee M, Na K, Hwang HS. Stem Cell-Derived Extracellular Vesicles for Cancer Therapy and Tissue Engineering Applications. Mol Pharm 2023; 20:5278-5311. [PMID: 37867343 DOI: 10.1021/acs.molpharmaceut.3c00376] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Recently, stem cells and their secretomes have attracted great attention in biomedical applications, particularly extracellular vesicles (EVs). EVs are secretomes of cells for cell-to-cell communication. They play a role as intercellular messengers as they carry proteins, nucleic acids, lipids, and therapeutic agents. They have also been utilized as drug-delivery vehicles due to their biocompatibility, low immunogenicity, stability, targetability, and engineerable properties. The therapeutic potential of EVs can be further enhanced by surface engineering and modification using functional molecules such as aptamers, peptides, and antibodies. As a consequence, EVs hold great promise as effective delivery vehicles for enhancing treatment efficacy while avoiding side effects. Among various cell types that secrete EVs, stem cells are ideal sources of EVs because stem cells have unique properties such as self-renewal and regenerative potential for transplantation into damaged tissues that can facilitate their regeneration. However, challenges such as immune rejection and ethical considerations remain significant hurdles. Stem cell-derived EVs have been extensively explored as a cell-free approach that bypasses many challenges associated with cell-based therapy in cancer therapy and tissue regeneration. In this review, we summarize and discuss the current knowledge of various types of stem cells as a source of EVs, their engineering, and applications of EVs, focusing on cancer therapy and tissue engineering.
Collapse
Affiliation(s)
- Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Kun Na
- Department of BioMedical-Chemical Engineering, The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
50
|
Yoo S, Choi S, Kim I, Kim IS. Hypoxic regulation of extracellular vesicles: Implications for cancer therapy. J Control Release 2023; 363:201-220. [PMID: 37739015 DOI: 10.1016/j.jconrel.2023.09.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Extracellular vesicles (EVs) play a pivotal role in intercellular communication and have been implicated in cancer progression. Hypoxia, a pervasive hallmark of cancer, is known to regulate EV biogenesis and function. Hypoxic EVs contain a specific set of proteins, nucleic acids, lipids, and metabolites, capable of reprogramming the biology and fate of recipient cells. Enhancing the intrinsic therapeutic efficacy of EVs can be achieved by strategically modifying their structure and contents. Moreover, the use of EVs as drug delivery vehicles holds great promise for cancer treatment. However, various hurdles must be overcome to enable their clinical application as cancer therapeutics. In this review, we aim to discuss the current knowledge on the hypoxic regulation of EVs. Additionally, we will describe the underlying mechanisms by which EVs contribute to cancer progression in hypoxia and outline the progress and limitations of hypoxia-related EV therapeutics for cancer.
Collapse
Affiliation(s)
- Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Sanga Choi
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea.
| |
Collapse
|