1
|
Chattopadhyay S, Rajendran RL, Chatterjee G, Reyaz D, Prakash K, Hong CM, Ahn BC, ArulJothi KN, Gangadaran P. Mesenchymal stem cell-derived exosomes: A paradigm shift in clinical therapeutics. Exp Cell Res 2025; 450:114616. [PMID: 40414452 DOI: 10.1016/j.yexcr.2025.114616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Mesenchymal stromal/stem cell (MSC)-derived exosomes are nanoscale extracellular vesicles that have emerged as promising candidates for therapeutic and diagnostic applications because of their unique bioactive cargo, including proteins, lipids, and nucleic acids. These vesicles mitigate concerns of immunogenicity and tumorigenicity associated with MSC-based therapies and offer enhanced stability, higher scalability, and ease of modification. However, the use of MSC-derived exosomes in clinical practice is associated with challenges, including difficulties in isolation, characterization, and standardization. This review explores the biogenesis and structural properties of MSC-derived exosomes and discusses the molecular mechanisms underlying their therapeutic effects. It also discusses ongoing clinical trials on their applications in cancer, cardiovascular, neurological, and regenerative medicine. Preclinical and clinical data have demonstrated the potential of MSC-derived exosomes in enhancing tissue repair, reducing inflammation, and modulating immune responses. Despite these advancements, gaps in scalable production methods, regulatory guidelines, and therapeutic consistency must be addressed. Future innovations in bioengineering, manufacturing, and regulatory frameworks are essential to realize the full potential of MSC-derived exosomes in mainstream medicine.
Collapse
Affiliation(s)
- Sayantani Chattopadhyay
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu, 603203, India
| | - Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Gargii Chatterjee
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu, 603203, India
| | - Danyal Reyaz
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu, 603203, India
| | - Kruthika Prakash
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu, 603203, India
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea; Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea.
| | - Kandasamy Nagarajan ArulJothi
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamilnadu, 603203, India.
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
2
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [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: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
3
|
Xiu X, Chen S, Liu Y, Sun B, Li H, Zhang S, Yang X, Wei Y, Peng X, Wang Y, Wang Y, Wu J, Zhang Y, Mu L, Kong Q, Liu X. Synergistic potential of bone marrow mesenchymal stem cells and miR181-a combinational therapy against multiple sclerosis. Stem Cell Res Ther 2025; 16:300. [PMID: 40490789 DOI: 10.1186/s13287-025-04401-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 05/16/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a progressive autoimmune disease characterized by massive inflammatory infiltration, demyelination, and subsequent axonal injury and neuronal damage in the central nervous system (CNS). The etiology of MS remains unclear and there is not yet a definitive therapeutic schedule for the disease. Bone marrow mesenchymal stem cells (BMSCs), exhibiting neuroimmune-modulatory functions to alleviate various autoimmune diseases, show great potential in the treatment of MS. However, the instability of BMSCs-mediated immunosuppression in vivo has limited their application. MiR181-a, a positive regulator of immune balance, which has a preference for T cells and B cells differentiation, but degrade rapidly upon entering systemic circulation due to their unstable molecular structure. METHODS We propose a synergistic therapy approach that combines the penetrative targeting capability of BMSCs with the immuno-modulatory effects of miR181-a by overexpressing miR181-a to BMSCs through lentivirus packaging system. With this strategy, on the basis of the establishment of the experimental autoimmune encephalomyelitis (EAE) model, miR181-a overexpressing BMSCs (miR181a-BMSCs) would have a stronger immuno-modulatory treatment benefit, in terms of attenuating MS development. RESULTS Indicate that this method prolongs the modulatory effects of BMSCs and resulted in significantly enhancements of the proliferation of regulatory B cells (Bregs), regulatory T cells (Tregs) and the inhibition of Th17 cells compared to the traditional BMSCs group. Moreover, 10-fold miRNA's concentration in the exosome of miR181a-BMSCs, leading to an increased duration of miRNAs to exert their biological effects. By immunotherapy and synergistic treatment, the effectiveness of the treatment is significantly enhanced, showing consistent results in different groups of the animal model. CONCLUSIONS This strategy takes advantage of BMSCs and miRNA and thus presents an effective synergistic strategy for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Xin Xiu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, 150081, China
| | - Sijia Chen
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yumei Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Bo Sun
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Hulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Sifan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xixi Yang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yu Wei
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xichen Peng
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yan Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yanping Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Junfeng Wu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yao Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Lili Mu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Qingfei Kong
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Xijun Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, 150081, China.
| |
Collapse
|
4
|
Zhu Q, Chen R, Wu X, Zhou Y, Wang Z, Zhang H, Zhu H, Sun L, Shuai Z. Bioinspired exosome-SiO 2 nanohybrid therapeutic for rheumatoid arthritis treatment. Theranostics 2025; 15:6553-6571. [PMID: 40521182 PMCID: PMC12160022 DOI: 10.7150/thno.108296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 05/11/2025] [Indexed: 06/18/2025] Open
Abstract
Objective: Due to their anti-inflammatory and immunomodulatory capabilities, adipose-derived stem cells (ADSC) are currently considered a promising option for the management of rheumatoid arthritis (RA). To tackle the problems of immunogenicity and tumorigenicity linked to the direct use of cells, current research is focused on the development of effective nanomedicines utilizing ADSC-derived exosomes (ADSC-EXO) for cell-free regenerative medicine. Methods: Methotrexate (MTX) was loaded into mesoporous silica through physical adsorption to produce SiO2-MTX, with subsequent incorporation into ADSC-EXO via ultrasonication to produce AE@SiO2-MTX. Particle size, surface charge, and stability were characterized using dynamic light scattering (DLS) and zeta potential analysis. In vitro, the effects of the nanomaterials were evaluated by assessing the inverse polarization effect of AE@SiO2-MTX on RAW264.7 macrophages, as well as on the migration and invasion capabilities of fibroblast-like synovial cells (FLS). In vivo, targeting and therapeutic effects on joint inflammation were examined using adjuvant-induced arthritis (AIA) and collagen-induced arthritis (CIA) mouse models. Results: The AE@SiO₂-MTX demonstrated sustained drug release, high biocompatibility, and rapid cellular internalization. In vitro, the delivery system alleviated chronic inflammation by inducing macrophage polarization from the pro-inflammatory M1 to the anti-inflammatory M2 phenotype, as well as suppressing FLS migration and invasion. In vivo studies revealed that administration of ADSC-EXO outperformed ADSC transplantation in alleviating RA symptoms. Intravenously delivered AE@SiO₂-MTX exhibited targeted accumulation in inflamed joints, significantly reducing joint swelling, synovial hyperplasia, and bone/cartilage degradation in CIA model mice. Conclusions: The findings show that AE@SiO₂-MTX is a robust cell-free therapeutic platform for RA management. Synergy between the immunomodulatory properties of ADSC-EXO and MTX controlled release, this system can overcome the limitations of conventional cell therapies and achieve targeted anti-inflammatory and tissue-protective effects. This strategy offers a promising translational avenue for RA treatment.
Collapse
Affiliation(s)
- Qicui Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ruofei Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xueting Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yuanyuan Zhou
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zexin Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Huaixuan Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Haofang Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| |
Collapse
|
5
|
Wang S, Wu J, Ren K, Zhang Y, Gao F, Chen Y, Chen C, Lu J. Platelet-Rich Plasma-Derived Exosome-Encapsulated Hydrogels Accelerate Diabetic Wound Healing by Inhibiting Fibroblast Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27923-27936. [PMID: 40315047 DOI: 10.1021/acsami.5c02705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Platelet-rich plasma-derived exosomes (PRP-Exos) have recently been considered an optimized strategy for diabetic wound treatment, yet the potential role of PRP-Exos in diabetic wound healing is still unclear. This study aims to investigate the potential mechanisms of PRP-Exos in diabetic wound healing and to utilize the hydrogel Pluronic F127 as a carrier to maintain the sustained release of encapsulated PRP-Exos. PRP-Exos were isolated from the blood of healthy individuals and characterized, followed by co-culturing with isolated diabetic human skin fibroblasts (Diabetes HSF). RNA sequencing (RNA-seq) was used to analyze the effect of PRP-Exos on the transcriptome of normal and Diabetes HSF, screening and validating the crucial mechanism and target gene. Then, a hydrogel composed of Pluronic F127 and PRP-Exos (PRP-Exos/Gel) was constructed and applied in the diabetic mouse models to evaluate the effect and mechanism. RNA-seq analysis revealed that PRP-Exos significantly upregulated the expression of FosB in Diabetes HSF. Further intervention in the expression of FosB in Diabetes HSF showed that knocking down FosB induced ferroptosis in Diabetes HSF, characterized by decreased cell viability, increased oxidative stress, and increased iron ion levels, along with downregulation of GPX4 and SLC7A11 expression, while ACSL4 expression was increased; conversely, overexpression of FosB had the opposite effect. Subsequently, adding PRP-Exos to FosB-knocked down Diabetes HSF significantly weakened the inhibitory effect of PRP-Exos on ferroptosis in diabetic fibroblasts. The synthesized PRP-Exos/Gel exhibited significant thermosensitivity and sustained release of exosomes. In animal experiments, the PRP-Exos/Gel showed significant anti-inflammatory effects, evidenced by an increased proportion of M2 macrophages and a decreased proportion of central granule cells in wound tissue, and inhibited fibroblast ferroptosis, thereby accelerating wound healing. Overall, the constructed PRP-Exos/Gel displays a continuous release of exosomes and promotes diabetic wound healing by suppressing inflammatory responses and fibroblast ferroptosis, which provides new insights and methods for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Shanzheng Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Jianyue Wu
- Department of Orthopaedics, Xishan People's Hospital of Wuxi, Wuxi, Jiangsu 214105, PR China
| | - Ke Ren
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Yuehou Zhang
- Department of Burn and Plastic Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Feifei Gao
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214400, PR China
| | - Yaofei Chen
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214400, PR China
| | - Changhong Chen
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214400, PR China
| | - Jun Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| |
Collapse
|
6
|
Sadeghi A, Noorbakhshnia M, Khodashenas S. Protective potential of BM-MSC extracted Exosomes in a rat model of Alzheimer's disease. PLoS One 2025; 20:e0320883. [PMID: 40327601 PMCID: PMC12054907 DOI: 10.1371/journal.pone.0320883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 02/25/2025] [Indexed: 05/08/2025] Open
Abstract
Exosomes are extracellular vesicles, which are released into the extracellular space by all types of cells, especially stem cells. Compared with stem cells, exosomes are safer and can be considered one of the most promising therapeutic strategies for neurodegenerative disease. We examined the effect of exosomes derived from bone marrow mesenchymal stem cells (BM-MSC) on a rat model of Alzheimer's disease (AD). For this purpose, male Wistar rats weighing 220-250 g were used. For the induction of AD, rats received a daily dosage of 100 mg/kg Aluminum chloride (Alcl3) by oral gavage for 60 days. Also, Primary BM-MSC was extracted from the femora of Wistar rats (male, 100-150 g). Extracted exosomes were Characterized and Qualified using TEM Microscope and Zetasizer Nano. Specific markers of exosomes were evaluated by Flow cytometry. MSC-extracted exosomes (150 µg/µl) were injected 2 or 5 times into the animals via tail vein on specific days. Our data revealed that receiving exosomes significantly prevented AlCl3-induced enhancement of hippocampal APP gene expression, beta-amyloid plaque formation, impairment of passive avoidance learning and spatial memory. However, exosome injections in healthy subjects caused some negative effects such as spatial memory impairment. It seems, MSC-derived exosomes can be considered as a candidate to prevent AD progression.
Collapse
Affiliation(s)
- Atefeh Sadeghi
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Maryam Noorbakhshnia
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Shabanali Khodashenas
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Thalassemia Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
7
|
Park S, Yoon YJ, Hong Y, Yu J, Cho JM, Jeong YJ, Yu H, Jeong H, Lee H, Hwang S, Koh WG, Yang JY, Hyun KA, Jung HI, Lim JY. CD9-enriched extracellular vesicles from chemically reprogrammed basal progenitors of salivary glands mitigate salivary gland fibrosis. Bioact Mater 2025; 47:229-247. [PMID: 39925710 PMCID: PMC11803853 DOI: 10.1016/j.bioactmat.2025.01.019] [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: 04/10/2024] [Revised: 12/07/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025] Open
Abstract
Extracellular vesicles (EVs) derived from stem cells offer promising potential for cell-free therapy. However, refining their cargo for precise disease targeting and delivery remains challenging. This study employed chemical reprogramming via dual inhibition of transforming growth factor beta (TGFβ) and bone morphogenetic protein (BMP) to expand salivary gland basal progenitor cells (sgBPCs). CD9-enriched (CD9+) EVs were then isolated from the sgBPC secretome concentrate using a dual microfluidic chip. Notably, CD9+ EVs demonstrated superior uptake by salivary epithelial cells compared to CD9-depleted (CD9-) EVs and total EVs. In vivo studies using a salivary gland (SG) obstruction mouse model and ex vivo studies in SG fibrosis organoids revealed that CD9+ EVs significantly enhanced anti-fibrotic effects over CD9- EVs and control treatments. The presence of miR-3162 and miR-1290 in CD9+ EVs supported their anti-fibrotic properties by downregulating ACVR1 expression. The chemical reprogramming culture method effectively expanded sgBPCs, enabling consistent and scalable EV production. Utilizing microfluidic chip-isolated CD9+ EVs and ductal delivery presents a targeted and efficient approach for anti-fibrotic SG regeneration.
Collapse
Affiliation(s)
- Sunyoung Park
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Yeo-Jun Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yongpyo Hong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jianning Yu
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- Department of Biomedical Laboratory Science, Yonsei University, 1 Yeonsedae-gil, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Jae-Min Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ye Jin Jeong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Haeun Yu
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Hyorim Jeong
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Hyunjin Lee
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Seungyeon Hwang
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Ji Yeong Yang
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Kyung-A Hyun
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- Korea Electronics Technology Institute (KETI), Seongnam, Gyeonggi-do, 13509, Republic of Korea
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
8
|
Li Z, Wang H, Zhou L, Chen C, Zheng X, Zhou C. The effect of electric field microenvironment on the exosome secretion of PC12 cells by chitosan carbon dots. Int J Biol Macromol 2025; 310:142747. [PMID: 40180068 DOI: 10.1016/j.ijbiomac.2025.142747] [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: 01/23/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
When neurons are sufficiently stimulated, they generate electrical signals with rapid voltage changes, and information is transferred between neurons through neurites. Chitosan carbon dots (CS CDs) possess unique optical properties, excellent biocompatibility, and superior electron transfer capabilities, making them promising materials for nerve tissue repair and replacement. Studies have shown that exosome (EXO) secretion by PC12 cells and neurite formation are critical for neural tissue repair. However, the effects of CDs on EXO secretion and neurite formation in an electric field microenvironment remain poorly explored. This study utilized CS to synthesize CDs and examined their impact on EXO secretion by PC12 cells in an electric field microenvironment. Findings demonstrated that CS-citric acid (CA) CDs had an average particle size of 4.5 nm with a lattice spacing of 0.248 nm and primarily consisted of graphitized carbon nuclei, hydroxyl, and amino groups. CS-CA CDs played a critical role in facilitating neurite elongation, especially with an electric field intensity of about 50 V/m. The combination of CS-CA CDs and electric field enhanced GAP43 expression in EXO secreted by PC12 cells. These results suggest that CS CDs have great potential for nerve injury treatment, repair, and regeneration.
Collapse
Affiliation(s)
- Zhuojuan Li
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, PR China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, PR China
| | - Lin Zhou
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, PR China
| | - Chengzhi Chen
- School of Life Science, Zhuhai College of Science and Technology, Zhuhai 519040, China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, 510630 Guangzhou, PR China.
| | - Changren Zhou
- School of Life Science, Zhuhai College of Science and Technology, Zhuhai 519040, China.
| |
Collapse
|
9
|
Kim M, Woo J, Kim J, Choi M, Shin HJ, Kim Y, Kim J, Shin DW. Iris germanica L. Rhizome-Derived Exosomes Ameliorated Dihydrotestosterone-Damaged Human Follicle Dermal Papilla Cells Through the Activation of Wnt/β-Catenin Pathway. Int J Mol Sci 2025; 26:4070. [PMID: 40362310 PMCID: PMC12071258 DOI: 10.3390/ijms26094070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Hair loss is often associated with oxidative stress and mitochondrial dysfunction in human follicle dermal papilla cells (HFDPCs), resulting in impaired cellular function and follicle degeneration. Thus, many studies have been conducted on natural plants aimed at inhibiting hair loss. This study investigated the therapeutic potential of exosomes derived from the rhizomes of Iris germanica L. (Iris-exosomes) in HFDPCs damaged by dihydrotestosterone (DHT). Iris-exosomes significantly reduced reactive oxygen species (ROS) levels, restoring mitochondrial membrane potential and ATP production, thereby mitigating oxidative stress and improving mitochondrial function. These effects occurred alongside enhanced cellular processes critical for hair follicle regeneration, including increased cell migration, alkaline phosphatase (ALP) activity, and three-dimensional (3D) spheroid formation, which replicates the follicle-like microenvironment and promotes inductive potential. Furthermore, Iris-exosomes stimulated the Wnt/β-catenin signaling pathway by enhancing glycogen synthase kinase-3β (GSK-3β), AKT, and extracellular signal-regulated kinase (ERK), leading to β-catenin stabilization and nuclear translocation, thereby supporting the expression of genes essential for hair growth. Taken together, these findings suggest that Iris-exosomes can be promising ingredients for alleviating hair loss.
Collapse
Affiliation(s)
- Mujun Kim
- Research Institute for Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; (M.K.); (J.K.)
| | - Jung Woo
- Shinsegae International Inc., Seoul 06015, Republic of Korea; (J.W.); (M.C.); (H.J.S.); (Y.K.); (J.K.)
| | - Jinsick Kim
- Research Institute for Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; (M.K.); (J.K.)
| | - Minah Choi
- Shinsegae International Inc., Seoul 06015, Republic of Korea; (J.W.); (M.C.); (H.J.S.); (Y.K.); (J.K.)
| | - Hee Jung Shin
- Shinsegae International Inc., Seoul 06015, Republic of Korea; (J.W.); (M.C.); (H.J.S.); (Y.K.); (J.K.)
| | - Youngseok Kim
- Shinsegae International Inc., Seoul 06015, Republic of Korea; (J.W.); (M.C.); (H.J.S.); (Y.K.); (J.K.)
| | - Junoh Kim
- Shinsegae International Inc., Seoul 06015, Republic of Korea; (J.W.); (M.C.); (H.J.S.); (Y.K.); (J.K.)
| | - Dong Wook Shin
- Research Institute for Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea; (M.K.); (J.K.)
| |
Collapse
|
10
|
Zhang Y, Yue NN, Chen LY, Tian CM, Yao J, Wang LS, Liang YJ, Wei DR, Ma HL, Li DF. Exosomal biomarkers: A novel frontier in the diagnosis of gastrointestinal cancers. World J Gastrointest Oncol 2025; 17:103591. [PMID: 40235899 PMCID: PMC11995328 DOI: 10.4251/wjgo.v17.i4.103591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Gastrointestinal (GI) cancers, which predominantly manifest in the stomach, colorectum, liver, esophagus, and pancreas, accounting for approximately 35% of global cancer-related mortality. The advent of liquid biopsy has introduced a pivotal diagnostic modality for the early identification of premalignant GI lesions and incipient cancers. This non-invasive technique not only facilitates prompt therapeutic intervention, but also serves as a critical adjunct in prognosticating the likelihood of tumor recurrence. The wealth of circulating exosomes present in body fluids is often enriched with proteins, lipids, microRNAs, and other RNAs derived from tumor cells. These specific cargo components are reflective of processes involved in GI tumorigenesis, tumor progression, and response to treatment. As such, they represent a group of promising biomarkers for aiding in the diagnosis of GI cancer. In this review, we delivered an exhaustive overview of the composition of exosomes and the pathways for cargo sorting within these vesicles. We laid out some of the clinical evidence that supported the utilization of exosomes as diagnostic biomarkers for GI cancers and discussed their potential for clinical application. Furthermore, we addressed the challenges encountered when harnessing exosomes as diagnostic and predictive instruments in the realm of GI cancers.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
- Department of Medical Administration, Huizhou Institute for Occupational Health, Huizhou 516000, Guangdong Province, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen 518000, Guangdong Province, China
| | - Li-Yu Chen
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (Jinan University of Second Clinical Medical Sciences), Shenzhen 518000, Guangdong Province, China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen 518000, Guangdong Province, China
| | - Dao-Ru Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Hua-Lin Ma
- Department of Nephrology, The Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong Province, China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| |
Collapse
|
11
|
Zi Y, Li J, Qian X, Li J, Jin Y, Zhang Z, Jin Y. Human umbilical cord mesenchymal stem cell exosomes promote elastin production and acute skin wound healing via TGFβ1-Smad pathway. Mol Cell Biochem 2025:10.1007/s11010-025-05264-5. [PMID: 40202710 DOI: 10.1007/s11010-025-05264-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Skin wound healing is a complex physiological process influenced by multiple factors, including the patient's overall health status. Exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSC-Exos) have demonstrated significant potential in enhancing wound repair. This study investigates the mechanisms through which hUCMSC-Exos facilitate skin wound healing and evaluates their potential application in combination with hydrogels for clinical treatment. Human foreskin fibroblasts (HFF-1) were treated with varying concentrations of hUCMSC-Exos to evaluate their impact on cell proliferation, assessed via the CCK-8 assay. Exosome uptake by HFF-1 cells was visualized using PKH-26 dye staining, while flow cytometry was employed to analyze cell cycle changes. Cell migration was evaluated through scratch and Transwell assays. Gene expression levels of Collagen I, Elastin, and Fibronectin were quantified by qRT-PCR, while Elastin secretion was measured by ELISA. Western blotting was used to examine proteins in the TGFβ1-Smad signaling pathway. The role of SP1 in regulating Elastin gene expression was investigated by testing the SP1 inhibitor Plicamycin and examining hUCMSC-Exos ability to counteract its effect. Additionally, a chromatin immunoprecipitation (ChIP) assay was performed to analyze SP1 binding at the Elastin gene promoter. In vivo, the efficacy of hUCMSC-Exos combined with hydrogels in promoting wound healing was assessed using a mouse skin wound model. hUCMSC-Exos significantly enhanced HFF-1 cell proliferation at concentrations exceeding 1 × 10⁹ particles/mL and increased the proportion of cells in the S and G2/M phases. HFF-1 cells readily absorbed these exosomes, leading to improved cell migration. Treatment with hUCMSC-Exos upregulated the gene expression of Collagen I, Fibronectin, and Elastin. The SP1 inhibitor Plicamycin reduced Elastin gene expression, an effect that was reversed by hUCMSC-Exos. In vivo, the combination of hUCMSC-Exos and hydrogels accelerated wound healing, enhanced collagen organization, and promoted the formation of elastic fibers and blood vessels. hUCMSC-Exos facilitate skin wound healing by promoting SP1 binding to the Elastin gene promoter, thereby upregulating Elastin expression and supporting extracellular matrix remodeling. These findings suggest a promising therapeutic role for hUCMSC-Exos in clinical applications for wound healing.
Collapse
Affiliation(s)
- Yi Zi
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - Jie Li
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - XinPing Qian
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - Jian Li
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - Yan Jin
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - ZiBo Zhang
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - YanHua Jin
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China.
| |
Collapse
|
12
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2025; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [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: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
13
|
He J, He F, Yang Q, Li Q. Blockade of Exosome Release Sensitizes Breast Cancer to Doxorubicin via Inhibiting Angiogenesis. Cancer Med 2025; 14:e70785. [PMID: 40249328 PMCID: PMC12007427 DOI: 10.1002/cam4.70785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Chemotherapy combined with angiogenesis inhibition holds great promise in improving the therapeutic efficacy in cancer treatment. The aim of this study was to explore the effect of exosome blockade on tumor angiogenesis and chemotherapy efficacy. METHODS Exosomes were extracted by ultracentrifugation, and the effect of exosomes on angiogenesis was evaluated by 4T1 mouse breast cancer cell line and the syngeneic mouse tumor model and immunofluorescence. The endocytosis of exosomes from vascular endothelial cells was evaluated in vitro by co-culture and immunofluorescence assays. Tube formation and CCK-8 assays were used to evaluate the effect of exosomes on angiogenesis in vitro. The effect of exosome blockade on the efficacy of doxorubicin was evaluated by 4T1 mouse breast cancer model, cancer cell-derived exosomes (Exo4T1), GW4869 and doxorubicin in vivo. RESULTS Exo4T1 can be efficiently endocytosed by vascular endothelial cells both in vitro and in vivo. Within the recipient endothelial cells, Exo4T1 elicited angiogenesis at least partially via promoting cell proliferation, as the exosomes were carrying cargos with pro-proliferation capacity. Blockade of exosome release through GW4869 significantly inhibited angiogenesis, increased the concentration of doxorubicin within the tumor, and sensitized the tumor to doxorubicin in the murine 4T1 syngeneic model, whereas the therapeutic effects were abrogated when Exo4T1 was additionally treated. Moreover, we found there was no synergy between GW4869 and pazopanib (PP, a traditional angiogenesis inhibitor). CONCLUSIONS Together, we here revealed that cancer-derived exosomes promote angiogenesis during cancer progression and GW4869 treatment would sensitize the cancer cells to doxorubicin at least partially via inhibiting angiogenesis.
Collapse
Affiliation(s)
- Jindi He
- Department of Breast SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Fengyi He
- Department of Breast SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Qinlian Yang
- Department of Breast SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| | - Qiuyun Li
- Department of Breast SurgeryGuangxi Medical University Cancer HospitalNanningGuangxiChina
| |
Collapse
|
14
|
Liu G, Cao R, Liu Q, Li H, Yan P, Wang K, Tian R, Yang P. M2 macrophages-derived exosomes for osteonecrosis of femoral head treatment: modulating neutrophil extracellular traps formation and endothelial phenotype transition. Bone Res 2025; 13:42. [PMID: 40169566 PMCID: PMC11961764 DOI: 10.1038/s41413-025-00412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 04/03/2025] Open
Abstract
Exosomes have shown good potential in ischemic injury disease treatments. However, evidence about their effect and molecular mechanisms in osteonecrosis of femoral head (ONFH) treatment is still limited. Here, we revealed the cell biology characters of ONFH osteonecrosis area bone tissue in single cell scale and thus identified a novel ONFH treatment approach based on M2 macrophages-derived exosomes (M2-Exos). We further show that M2-Exos are highly effective in the treatment of ONFH by modulating the phenotypes communication between neutrophil and endothelium including neutrophil extracellular traps formation and endothelial phenotype transition. Additionally, we identified that M2-Exos' therapeutic effect is attributed to the high content of miR-93-5p and constructed miR-93-5p overexpression model in vitro and in vivo based on lentivirus and adeno-associated virus respectively. Then we found miR-93-5p can not only reduce neutrophil extracellular traps formation but also improve angiogenic ability of endothelial cells. These results provided a new theoretical basis for the clinical application of ONFH therapeutic exosomes.
Collapse
Affiliation(s)
- Guanzhi Liu
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruomu Cao
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qimeng Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Li
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peng Yan
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kunzheng Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Run Tian
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pei Yang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
15
|
Daksh R, Mathew MS, Bosco AM, Sojan C, Tom AA, Bojja SL, Nampoothiri M. The role of exosomes in diagnosis, pathophysiology, and management of Alzheimer's Disease. Biochem Biophys Res Commun 2025; 754:151526. [PMID: 40015072 DOI: 10.1016/j.bbrc.2025.151526] [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: 01/12/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with impaired cognitive function and memory loss. Currently, available therapeutics can effectively alleviate the symptoms of AD, but there is a lack of treatment to halt the progression of the disease. In recent years, exosomes have gained much attention due to their involvement in various neurological disorders. Exosomes are small extracellular vesicles comprising lipids, proteins, DNA, non-coding RNA, and mRNAs, can carry various therapeutic molecules, and are potential drug delivery vehicles. Exosomes are known as a double-edged sword due to their involvement in both the pathogenesis and management of AD. This review explores the function of exosomes in the pathophysiology, treatment, and diagnosis of AD, also emphasizing their potential as a targeted drug delivery carrier to the brain. This review seeks to provide novel perspectives to understand better the onset, targeted treatment, and diagnosis of AD using exosomes.
Collapse
Affiliation(s)
- Rajni Daksh
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Meby Susan Mathew
- Nirmala College of Pharmacy, Kerala University of Health Sciences, Kerala, India
| | - Aan Mery Bosco
- Nirmala College of Pharmacy, Kerala University of Health Sciences, Kerala, India
| | - Christy Sojan
- Nirmala College of Pharmacy, Kerala University of Health Sciences, Kerala, India
| | - Antriya Annie Tom
- Nirmala College of Pharmacy, Kerala University of Health Sciences, Kerala, India
| | - Sree Lalitha Bojja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
16
|
Wang W, Wang J, Liao D. Effects and Mechanisms of Extracellular Vesicles in Different Models of Acute Kidney Injury. Stem Cells Int 2025; 2025:1075016. [PMID: 40165854 PMCID: PMC11957863 DOI: 10.1155/sci/1075016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Acute kidney injury (AKI) is a rapid decline in renal function caused by ischemia/reperfusion (I/R), renal toxic injury, and sepsis. While the precise molecular mechanisms underlying AKI are still under investigation, current therapeutic approaches remain insufficient. In recent years, there has been growing evidence that mesenchymal stem cells (MSCs) have great potential in accelerating renal repair after AKI in various preclinical models, while there has been extensive research on extracellular vesicles (EVs) as therapeutic mediators in AKI models, and they are considered to be superior to MSCs as new regenerative therapies. EVs are nanoparticles secreted by various types of cells under physiological and pathological conditions. EVs derived from various sources possess biomarker potential and play crucial roles in mediating cellular communication between kidney cells and other tissue cells by transmitting signal molecules. These vesicles play a direct and indirect role in regulating the pathophysiological mechanisms of AKI and contribute to the occurrence, development, treatment, and repair of AKI. In this review, we briefly outline the essential characteristics of EVs, focus on the multiple molecular mechanisms currently involved in the protection of EVs against different types of AKI, and further discuss the potential targets of EVs from different sources in the treatment of AKI. Finally, we summarized the deficiencies in the production and treatment of EVs and the current strategies for improvement.
Collapse
Affiliation(s)
- Weidong Wang
- Department of Nephrology, Mianyang Central Hospital, Mianyang 621000, China
| | - Jingyu Wang
- Renal Division, Peking University First Hospital, Beijing 100080, China
| | - Dan Liao
- Department of Nephrology, Mianyang Central Hospital, Mianyang 621000, China
| |
Collapse
|
17
|
Ai S, Xie Z, Li N, Zhao R, Qu X, Zhou H, Tang D, Zhang J, Luo X. Bibliometric and visualized analysis of the applications of exosomes for bone regeneration. Front Cell Dev Biol 2025; 13:1552727. [PMID: 40166633 PMCID: PMC11955700 DOI: 10.3389/fcell.2025.1552727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Background Bone defect, a common orthopedic condition, is characterized by a lengthy and impactful treatment period, posing a considerable challenge in clinical settings. Medical technology has advanced notably, and has effectively treated an increasing number of patients with bone defects. Consequently, there has been an explosion of research articles on bone regeneration, including a substantial number on the application of exosomes. Exosomes, especially those derived from stem cells, have been confirmed to be effective in bone regeneration and have garnered widespread attention in the last decade. Therefore, this study conducted a bibliometric analysis on publications related to the application of exosomes for bone regeneration. The objectives are to explore the development history and research hotspots in this field over the past 10 years, predict future development trends, and provide guidance for subsequent research. Methods The Web of Science Core Collection (WoSCC) database was searched for articles related to exosomes and bone regeneration published from 1 January 2014, to 31 December 2023. The collected literature was analyzed using software such as Microsoft Excel, CiteSpace 6.3R1, VOSviewer 1.6.20, and the bibliometric online platform (https://bibliometric.com). Results A total of 3,004 articles published by 2,729 institutions from 68 countries were included in this study. The number of articles on the application of exosomes for bone regeneration has increased annually over the last decade. China was the most prolific country in this field, with a total of 1,468 papers; Shanghai Jiao Tong University (China) was the institution with the highest number of publications (117 publications). In terms of authors, Xin Wang, Yi Zhang, and Yang Wang were the three who published the highest number of papers, with 14 papers each. Co-citation analysis revealed that the article published by Valadi H in 2007 has the highest number of co-citations (270 times of quotation). Additionally, most research hotspots focused on the function of exosomes and the mechanism of action. Furthermore, the importance of osteoblast differentiation and angiogenesis in bone regeneration has also garnered significant attention from scholars in this field. Conclusion This study reviewed the research achievements on the application of exosomes for bone regeneration over the past 10 years, utilizing bibliometric analysis tools. It visualized the countries, institutions, authors, and journals that have made significant contributions to this field, revealed current research hotspots, and finally explored future development trends.
Collapse
Affiliation(s)
- Shuai Ai
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Zhou Xie
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Ningdao Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Runhan Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Xiao Qu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Haining Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Dagang Tang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Jun Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
| | - Xiaoji Luo
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, China
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| |
Collapse
|
18
|
Abualigah L, Alomari SA, Almomani MH, Zitar RA, Saleem K, Migdady H, Snasel V, Smerat A, Ezugwu AE. Artificial intelligence-driven translational medicine: a machine learning framework for predicting disease outcomes and optimizing patient-centric care. J Transl Med 2025; 23:302. [PMID: 40065389 PMCID: PMC11892274 DOI: 10.1186/s12967-025-06308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Advancements in artificial intelligence (AI) and machine learning (ML) have revolutionized the medical field and transformed translational medicine. These technologies enable more accurate disease trajectory models while enhancing patient-centered care. However, challenges such as heterogeneous datasets, class imbalance, and scalability remain barriers to achieving optimal predictive performance. METHODS This study proposes a novel AI-based framework that integrates Gradient Boosting Machines (GBM) and Deep Neural Networks (DNN) to address these challenges. The framework was evaluated using two distinct datasets: MIMIC-IV, a critical care database containing clinical data of critically ill patients, and the UK Biobank, which comprises genetic, clinical, and lifestyle data from 500,000 participants. Key performance metrics, including Accuracy, Precision, Recall, F1-Score, and AUROC, were used to assess the framework against traditional and advanced ML models. RESULTS The proposed framework demonstrated superior performance compared to classical models such as Logistic Regression, Random Forest, Support Vector Machines (SVM), and Neural Networks. For example, on the UK Biobank dataset, the model achieved an AUROC of 0.96, significantly outperforming Neural Networks (0.92). The framework was also efficient, requiring only 32.4 s for training on MIMIC-IV, with low prediction latency, making it suitable for real-time applications. CONCLUSIONS The proposed AI-based framework effectively addresses critical challenges in translational medicine, offering superior predictive accuracy and efficiency. Its robust performance across diverse datasets highlights its potential for integration into real-time clinical decision support systems, facilitating personalized medicine and improving patient outcomes. Future research will focus on enhancing scalability and interpretability for broader clinical applications.
Collapse
Affiliation(s)
- Laith Abualigah
- Computer Science Department, Al Al-Bayt University, Mafraq, 25113, Jordan.
| | - Saleh Ali Alomari
- Faculty of Science and Information Technology, Jadara University, Irbid, 21110, Jordan
| | - Mohammad H Almomani
- Department of Mathematics, Facility of Science, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Raed Abu Zitar
- Faculty of Engineering and Computing, Liwa College, Abu Dhabi, United Arab Emirates
| | - Kashif Saleem
- Department of Computer Science and Engineering, College of Applied Studies and Community Service, King Saud University, 11362, Riyadh, Saudi Arabia
| | - Hazem Migdady
- CSMIS Department, Oman College of Management and Technology, 320, Barka, Oman
| | - Vaclav Snasel
- Faculty of Electrical Engineering and Computer Science, VŠB-Technical University of Ostrava, 70800, Poruba-Ostrava, Czech Republic
| | - Aseel Smerat
- Faculty of Educational Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Absalom E Ezugwu
- Unit for Data Science and Computing, North-West University, 11 Hofman Street, Potchefstroom, 2520, South Africa.
| |
Collapse
|
19
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
20
|
Sharma Y, Mohanty S. Targeted knockdown of MSC-sEVs biogenesis regulator proteins to elucidate the mechanisms of their production: a step towards translational applications. Cytotherapy 2025:S1465-3249(25)00061-1. [PMID: 39985543 DOI: 10.1016/j.jcyt.2025.02.002] [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: 11/20/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
In the intricate landscape of cellular communication, small extracellular vesicles (sEVs) originating from endosomes play crucial roles as mediators and have garnered significant attention in theranostics. Our understanding of sEV biogenesis largely stems from studies on cancer cells, which are vital for diagnostics. However, in therapeutics, where mesenchymal stromal cell (MSC)-derived sEVs are emerging as investigational new drugs, their biogenesis pathways remain largely unexplored. This article explores the parallel narratives of sEV biogenesis in cancer cells and stem cells, specifically using HeLa cells and MSCs as model cell lines. This study investigated the roles of key proteins-hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), signal-transducing adaptor molecule (STAM), tumor susceptibility gene 101 (TSG101), and ALG-2-interacting protein X (ALIX)-as identified in HeLa cells, in the context of MSC-sEV biogenesis. While these proteins show similarities across cell types, a discernible difference arises in their primary functions in regulating sEV biogenesis. The critical role of ALIX in MSC-sEV biogenesis, in particular, underscores its potential as a target for modulating sEVs' yield in regenerative therapies. Through this comparative analysis, we identified shared molecular signatures, offering insights to guide therapeutic interventions and unlock the regenerative potential of stem cells.
Collapse
Affiliation(s)
- Yashvi Sharma
- Stem Cell Facility-DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility-DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
21
|
Powantia Pontoh LA, Fiolin J, Dilogo IH, Prasetyo M, Antarianto RD, Harahap A, Tantry AJ, Pakasi TA, Priosoeryanto BP, Dewi TIT. Combined exosome of adipose-derived mesenchymal stem cell and hyaluronic acid delays early osteoarthritis progression of ovine sheep model: Clinical, radiographic, macroscopic and microscopic evaluation. F1000Res 2025; 13:494. [PMID: 39989481 PMCID: PMC11842963 DOI: 10.12688/f1000research.147309.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 02/25/2025] Open
Abstract
Background Current treatment of osteoarthritis (OA) mainly focused on treating symptoms. Exosome from Adipose-derived Mesenchymal Stem Cell (Ad-MSC) have been shown to delay degenerative process. This study aimed to investigate the clinical, radiological and histological impact of combined intra-articular (IA) hyaluronic acid (HA) and exosome Ad-MSCs in-vivo using a larger animal model with low-grade OA. Methods Eighteen male Ovies aries sheep underwent total lateral meniscectomy and conventional radiography was performed to confirm low-grade OA after 6 weeks. The sheep were divided into three groups, Group 1 (G1; n=6) received thrice exosome injections, G2 (n=6) received twice HA injection, and G3 (n=6) received both treatments with a 1-week interval after 10 days of meniscectomy. Clinical evaluations were conducted using the Clinical Lameness Score (CLS), radiographic with X-ray using OA score by Innes et al, while macroscopic evaluation by Osteoarthritis Research Society International (OARSI) scores. Results Lameness parameter scored lowest in G3 significantly (2.0±0.0 VS 2.7±0.52 VS 2.7±0.52; p=0.024) at the second month although the overall CLS score did not significantly differ at the 3 rd month. The best improvement of conventional total OA radiographic score at the 3 rd month compared to all groups (5.2±1.17 vs 6.3±0.82 vs 6.7±1.03; p=0.053). Macroscopic OARSI evaluation showed no difference (p=0.711). Conclusions Combined repeated exosome Ad-MSC and HA IA injection proven to delay OA progression, however longer duration of follow up is required to evaluate its long-term effect.
Collapse
Affiliation(s)
| | - Jessica Fiolin
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Indonesia, Central Jakarta, DKI Jakarta, 10430, Indonesia
| | - Ismail Hadisoebroto Dilogo
- Orthopaedic and Traumatology, Faculty of Medicine, Universitas Indonesia, Centra Jakarta, DKI Jakarta, 10430, Indonesia
| | - Marcel Prasetyo
- Department of Radiology, Faculty of Medicine, Universitas Indonesia, Central Jakarta, DKI Jakarta, 10430, Indonesia
| | - Radiana Dhewayani Antarianto
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Central Jakarta, DKI Jakarta, 10430, Indonesia
| | - Alida Harahap
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Indonesia, Central Jakarta, DKI Jakarta, 10430, Indonesia
| | - Angela Jennifer Tantry
- Department of Biology, Faculty of Mathematics and Natural Science, Universitas Indonesia, Depok, West Java, 16424, Indonesia
| | - Trevino Aristakus Pakasi
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Central Jakarta, DKI Jakarta, 10430, Indonesia
| | - Bambang Pontjo Priosoeryanto
- School of Veterinary Medicine and Biomedical Science, Institut Pertanian Bogor, Bogor, West Java, 16680, Indonesia
| | - Tri Isyani Tungga Dewi
- School of Veterinary Medicine and Biomedical Science, Institut Pertanian Bogor, Bogor, West Java, 16680, Indonesia
| |
Collapse
|
22
|
Lu Y, Chen T, Lin H, Chen Y, Lin Y, Le D, Huang Y, Wang AH, Lee C, Ling T. Small Extracellular Vesicles Engineered Using Click Chemistry to Express Chimeric Antigen Receptors Show Enhanced Efficacy in Acute Liver Failure. J Extracell Vesicles 2025; 14:e70044. [PMID: 39901768 PMCID: PMC11791321 DOI: 10.1002/jev2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 02/05/2025] Open
Abstract
Acetaminophen (APAP) overdose can cause severe liver injury and life-threatening conditions that may lead to multiple organ failure without proper treatment. N-acetylcysteine (NAC) is the accepted and prescribed treatment for detoxification in cases of APAP overdose. Nonetheless, in acute liver failure (ALF), particularly when the ingestion is substantial, NAC may not fully restore liver function. NAC administration in ALF has limitations and potential adverse effects, including nausea, vomiting, diarrhoea, flatus, gastroesophageal reflux, and anaphylactoid reactions. Mesenchymal stromal cell (MSC)-based therapies using paracrine activity show promise for treating ALF, with preclinical studies demonstrating improvement. Recently, MSC-derived extracellular vesicles (EVs) have emerged as a new therapeutic option for liver injury. MSC-derived EVs can contain various therapeutic cargos depending on the cell of origin, participate in physiological processes, and respond to abnormalities. However, most therapeutic EVs lack a distinct orientation upon entering the body, resulting in a lack of targeting specificity. Therefore, enhancing the precision of natural EV delivery systems is urgently needed. Thus, we developed an advanced targeting technique to deliver modified EVs within the body. Our strategy aims to employ bioorthogonal click chemistry to attach a targeting molecule to the surface of small extracellular vesicles (sEVs), creating exogenous chimeric antigen receptor-modified sEVs (CAR-sEVs) for the treatment. First, we engineered azido-modified sEVs (N3-sEVs) through metabolic glycoengineering by treating MSCs with the azide-containing monosaccharide N-azidoacetyl-mannosamine (Ac4ManNAz). Next, we conjugated N3-sEVs with a dibenzocyclooctyne (DBCO)-tagged single-chain variable fragment (DBCO-scFv) that targets the asialoglycoprotein receptor (ASGR1), thus producing CAR-sEVs for precise liver targeting. The efficacy of CAR-sEV therapy in ALF models by targeting ASGR1 was validated. MSC-derived CAR-sEVs reduced serum liver enzymes, mitigated liver damage, and promoted hepatocyte proliferation in APAP-induced injury. Overall, CAR-sEVs exhibited enhanced hepatocyte specificity and efficacy in ameliorating liver injury, highlighting the significant advancements achievable with cell-free targeted therapy.
Collapse
Affiliation(s)
- Yen‐Ting Lu
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Tzu‐Yu Chen
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Hsin‐Hung Lin
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
- MediDiamond Inc.TaipeiTaiwan
| | - Ya‐Wen Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Yu‐Xiu Lin
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Duy‑Cuong Le
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Vinmec Hi‐Tech CenterVinmec Healthcare SystemHanoiVietnam
- International Ph.D. Program for Translational Science, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Yen‐Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- TMU Research Center of Cell Therapy and Regeneration MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Andrew H.‐J. Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Cheng‐Chung Lee
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Ph.D. Program for Translational Science, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Thai‐Yen Ling
- Graduate Institute of PharmacologyNational Taiwan University College of MedicineTaipeiTaiwan
| |
Collapse
|
23
|
Tariq H, Bukhari SZ, An R, Dong J, Ihsan A, Younis MR. Stem cell-derived exosome delivery systems for treating atherosclerosis: The new frontier of stem cell therapy. Mater Today Bio 2025; 30:101440. [PMID: 39866781 PMCID: PMC11758955 DOI: 10.1016/j.mtbio.2024.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide. As a chronic inflammatory disease with a complicated pathophysiology marked by abnormal lipid metabolism and arterial plaque formation, atherosclerosis is a major contributor to CVDs and can induce abrupt cardiac events. The discovery of exosomes' role in intercellular communication has sparked a great deal of interest in them recently. Exosomes are involved in strategic phases of the onset and development of atherosclerosis because they have been identified to control pathophysiologic pathways including inflammation, angiogenesis, or senescence. This review investigates the potential role of stem cell-derived exosomes in atherosclerosis management. We briefly introduced atherosclerosis and stem cell therapy including stem cell-derived exosomes. The biogenesis of exosomes along with their secretion and isolation have been elaborated. The design engineering of exosomes has been summarized to present how drug loading and surface modification with targeting ligands can improve the therapeutic and targeting capacity of exosomes, demonstrating atheroprotective action. Moreover, the mechanism of action (endothelial dysfunction, reduction of dyslipidemia, macrophage polarization, vascular calcification, and angiogenesis) of drug-loaded exosomes to treat atherosclerosis has been discussed in detail. In the end, a comparative and balanced viewpoint has been given regarding the current challenges and potential solutions to advance exosome engineering for cardiovascular therapeutic applications.
Collapse
Affiliation(s)
- Hassan Tariq
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| | - Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ruibing An
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Jian Dong
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Rizwan Younis
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
24
|
Kim P, Joe S, Kim H, Jeong H, Park S, Song J, Kim W, Lee YG. Hidden Partner of Immunity: Microbiome as an Innovative Companion in Immunotherapy. Int J Mol Sci 2025; 26:856. [PMID: 39859572 PMCID: PMC11765694 DOI: 10.3390/ijms26020856] [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: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Recent studies have highlighted that the microbiome is the essential factor that can modulate the clinical activity of immunotherapy. However, the role of the microbiome varies significantly across different immunotherapies, suggesting that it is critical to understand the precise function of the microbiome in each type of immunotherapy. While many previous studies primarily focus on summarizing the role of the microbiome in immune checkpoint inhibitors, we seek to explore a novel aspect of the microbiome in other immunotherapies such as mesenchymal stem cell therapy, chimeric antigen receptor T cell therapy, and antibodies-based therapy (e.g., adalimumab, infliximab, bevacizumab, denosumab, etc.) which are rarely summarized in previous reviews. Moreover, we highlight innovative strategies for utilizing microbiome and microbial metabolites to enhance the clinical response of immunotherapy. Collectively, we believe that our manuscript will provide novel insights and innovative approaches to the researchers, which could drive the development of the next generation of personalized therapeutic interventions using microbiomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wondong Kim
- Correspondence: (W.K.); (Y.G.L.); Tel.: +82-31-400-5817 (W.K.); +82-31-400-5814 (Y.G.L.)
| | - Yong Gu Lee
- Correspondence: (W.K.); (Y.G.L.); Tel.: +82-31-400-5817 (W.K.); +82-31-400-5814 (Y.G.L.)
| |
Collapse
|
25
|
Yang F, Ni B, Liang X, He Y, Yuan C, Chu J, Huang Y, Zhong H, Yang L, Lu J, Xu Y, Zhang Q, Chen W. Mesenchymal stromal cell-derived extracellular vesicles as nanotherapeutics for concanavalin a-induced hepatitis: modulating the gut‒liver axis. Stem Cell Res Ther 2025; 16:4. [PMID: 39773662 PMCID: PMC11706160 DOI: 10.1186/s13287-024-04013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND As cell-free nanotherapeutics, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have shown potential therapeutic action against liver diseases. However, their effects on autoimmune hepatitis (AIH) are not yet well understood. METHODS AND RESULTS In this study, we utilized a well-established concanavalin A (Con A)-induced fulminant hepatitis mouse model to investigate the effects of MSC-EVs on AIH. We found that MSC-EVs provide significant protection against Con A-induced hepatitis in C57BL/6 male mice, with their effectiveness being critically dependent on the gut microbiota. MSC-EVs modulate the composition of the gut microbiota, particularly by increasing the abundance of norank_f__Muribaculaceae, and impact liver metabolic profiles, leading to significant amelioration of liver injury. The identification of Acetyl-DL-Valine as a protective metabolite underscores the therapeutic potential of targeting gut‒liver axis interactions in liver diseases. CONCLUSION Overall, our data demonstrate that MSC-EVs exhibit nanotherapeutic potential in Con A-induced hepatitis and provide new insights into the treatment of autoimmune hepatitis.
Collapse
Affiliation(s)
- Fan Yang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
- Xinjiang Stem Cells Special Plateau Disease Engineering Technology Research Center, The First People's Hospital of Kashi, The Affiliated Kashi Hospital of Sun Yat-Sen University), Kashi, 844000, P.R. China
| | - Beibei Ni
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
| | - Xiaoqi Liang
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
| | - Yizhan He
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University Zhaoqing hospital, Zhaoqing, 526070, P.R. China
| | - Chao Yuan
- General practice, Guangdong provincial people's hospital, Guangzhou, 510080, P.R. China
| | - Jiajie Chu
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
| | - Yiju Huang
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
| | - Hongyu Zhong
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
| | - Li Yang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
- Xinjiang Stem Cells Special Plateau Disease Engineering Technology Research Center, The First People's Hospital of Kashi, The Affiliated Kashi Hospital of Sun Yat-Sen University), Kashi, 844000, P.R. China
| | - Jianxi Lu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
- Xinjiang Stem Cells Special Plateau Disease Engineering Technology Research Center, The First People's Hospital of Kashi, The Affiliated Kashi Hospital of Sun Yat-Sen University), Kashi, 844000, P.R. China
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China
| | - Yan Xu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China.
- Xinjiang Stem Cells Special Plateau Disease Engineering Technology Research Center, The First People's Hospital of Kashi, The Affiliated Kashi Hospital of Sun Yat-Sen University), Kashi, 844000, P.R. China.
| | - Qi Zhang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China.
- Xinjiang Stem Cells Special Plateau Disease Engineering Technology Research Center, The First People's Hospital of Kashi, The Affiliated Kashi Hospital of Sun Yat-Sen University), Kashi, 844000, P.R. China.
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China.
| | - Wenjie Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China.
- Xinjiang Stem Cells Special Plateau Disease Engineering Technology Research Center, The First People's Hospital of Kashi, The Affiliated Kashi Hospital of Sun Yat-Sen University), Kashi, 844000, P.R. China.
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, P.R. China.
| |
Collapse
|
26
|
Wen Y, Liang Z. Enhanced itaconic acid secretion from macrophages mediates the protection of mesenchymal stem cell-derived exosomes on lipopolysaccharide-induced acute lung injury mice. Biol Direct 2024; 19:138. [PMID: 39721998 DOI: 10.1186/s13062-024-00586-8] [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: 08/12/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Alveolar macrophages (AMs) is critical to exacerbate acute lung injury (ALI) induced by lipopolysaccharide (LPS) via inhibiting inflammation, which could by shifted by mesenchymal stem cell-derived exosomes (MSC-exos). But the underlying rationale is not fully clarified. Our study aimed to analyze the significance of itaconic acid (ITA) in mediating the protective effects of MSC-exos on LPS-induced ALI. METHODS MSC-exos were used to treat pulmonary microvascular endothelial cells (PMVECs) co-cultured with AMs under LPS stimulation. si-IRG1 was transfected to AMs. PMVEC permeability, apoptosis rates, and inflammatory cytokine levels were assessed. In vivo, C57BL/6 wild-type (WT) and Irg1-/- mice were employed to explore the protection of MSC-exos against LPS-induced ALI. The lung injury was determined by histological and biochemical assays. ITA levels were measured using gas chromatography-mass spectrometry. Western blot and flow cytometry analyses were performed to assess M1/M2 polarization. RESULTS Co-culture with AMs significantly increased PMVEC permeability, apoptosis rates, IL-6, TNF-α levels and Claudin-5 and ZO-1 expression induced by LPS treatment, which were attenuated by MSC-exos accompanied by enhanced ITA level. After si-IRG1 transfection, MSC-exos' protective efficacy was reversed, with suppressed M2 polarization. In vivo, MSC-exos alleviated alveolar structure disruption, pulmonary edema, inflammation and increased ITA concentration in WT mice but had reduced effects in Irg1-/- mice, with neglected M2 polarization. CONCLUSIONS ITA secretion facilitated the MSC-exos' protective benefits on LPS-induced PMVEC damage and ALI in mice by promoting AM M2 polarization, highlighting a potential therapeutic strategy for ALI and related inflammatory lung diseases.
Collapse
Affiliation(s)
- Yanmei Wen
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, Chengdu Second People's Hospital, Chengdu, 610000, Sichuan, China
| | - Zong'an Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
27
|
Qu Q, Liu L, Wang L, Cui Y, Liu C, Jing X, Xu X. Exosomes derived from hypoxic mesenchymal stem cells restore ovarian function by enhancing angiogenesis. Stem Cell Res Ther 2024; 15:496. [PMID: 39709481 DOI: 10.1186/s13287-024-04111-6] [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: 05/10/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND hucMSC-exosomes can be engineered to strengthen their therapeutic potential, and the present study aimed to explore whether hypoxic preconditioning can enhance the angiogenic potential of hucMSC-exosomes in an experimental model of POF. METHODS Primary hucMSCs and ROMECs were isolated from fresh tissue samples and assessed through a series of experiments. Exosomes were isolated from hucMSCs under normoxic or hypoxic conditions (norm-Exos and hypo-Exos, respectively) and then characterized using classic experimental methods. Based on a series of angiogenesis-related assays, we found that hypo-Exos significantly promoted ROMEC proliferation, migration, and tube formation and increased angiogenesis-promoting molecules in vitro. Histology, immunohistochemistry, and immunofluorescence experiments in a rat model of POF demonstrated that hypoxia pretreatment strengthens the therapeutic angiogenic effect of hucMSC-exosomes in vivo. Subsequently, high-throughput miRNA sequencing, qRT‑PCR analysis, and western blotting were employed to identify the potential molecular mechanism. RESULTS We found that hypo-Exos enhance endothelial function and angiogenesis via the transfer of miR-205-5p in vitro and in vivo. Finally, based on the results of bioinformatics analysis, dual luciferase reporter assays, and gain- and loss-of-function studies, we found evidence indicating that exosomal miR-205-5p enhances angiogenesis by targeting the PTEN/PI3K/AKT/mTOR signalling pathway. These results indicated for the first time that exosomes derived from hypoxia-conditioned hucMSCs strongly enhance angiogenesis via the transfer of miR-205-5p by targeting the PTEN/PI3K/AKT/mTOR signalling pathway. CONCLUSIONS Our findings provide a theoretical basis and demonstrate the potential application of a novel cell-free approach with stem cell-derived products in the treatment of POF.
Collapse
Affiliation(s)
- Qingxi Qu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Linghong Liu
- Research Center of Stem Cell and Regenerative Medicine, Shandong University, Jinan, 250012, P.R. China.
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China.
| | - Limei Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Yuqian Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Chunxiao Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Xuanxuan Jing
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Xiaoxuan Xu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| |
Collapse
|
28
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [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: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
29
|
Hadizadeh A, Akbari Asbagh R, Heirani-Tabasi A, Soleimani M, Gorovanchi P, Ebrahimi Daryani N, Vahedi A, Nazari H, Banikarimi SP, Abbaszade Dibavar M, Behboudi B, Fazeli MS, Keramati MR, Keshvari A, Kazemeini A, Pak H, Fazeli AR, Alborzi Avanaki F, Ahmadi-Tafti SM. Localized Administration of Mesenchymal Stem Cell-Derived Exosomes for the Treatment of Refractory Perianal Fistula in Patients With Crohn's Disease: A Phase II Clinical Trial. Dis Colon Rectum 2024; 67:1564-1575. [PMID: 39250316 DOI: 10.1097/dcr.0000000000003502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
BACKGROUND Crohn's disease perianal fistulas are often resistant to standard anti-tumor necrosis factor-α therapies. Mesenchymal stem cell-derived exosomes are extracellular vesicles with highly potent anti-inflammatory effects, and the previous phase of this study demonstrated their safety in the treatment of refractory perianal fistulas. OBJECTIVE To evaluate the efficacy of mesenchymal stem cell-derived exosomes for the treatment of refractory perianal fistulas. DESIGN Nonrandomized, nonblinded single-center phase II clinical trial. SETTINGS Tertiary university hospital. PATIENTS Twenty-three patients were enrolled, 20 of whom completed the study. Refractory perianal fistula was defined as resistance to at least 1 course of treatment with anti-tumor necrosis factor-α therapy. INTERVENTIONS After clinical assessment and MRI, the patients received general anesthesia, and 5 mL of exosome solution was injected directly into the fistula tracts. The injections were repeated 3 times at 2-month intervals, and patients were followed monthly for 6 months after the last injection. Tissue samples from the tracts were obtained before each injection and subjected to immunohistopathological assessment. MRI data were obtained before and 6 months after the last injection. MAIN OUTCOME MEASURES The primary outcome of this study was fistula tract closure on clinical examination and MRI. The secondary outcome was an improvement in the discharge from the tracts. RESULTS Fistula tracts were fully closed in 12 patients (60%). Four patients showed clinical improvement, with some tracts remaining open, and 4 patients were completely resistant to treatment. A total of 43 fistula tracts were treated during the trial, 30 of which (69.7%) showed complete closure. Histopathological analysis revealed substantial reductions in local inflammation and signs of enhanced tissue regeneration. Immunohistochemical analysis of CD68, CD20, and CD31 reaffirmed these results. CONCLUSIONS Mesenchymal stem cell-derived exosomes are safe and effective for treating refractory perianal fistulas in patients with Crohn's disease. See Video Abstract . ADMINISTRACIN LOCALIZADA DE EXOSOMAS DERIVADOS DE CLULAS MADRE MESENQUIMALES PARA EL TRATAMIENTO DE LA FSTULA PERIANAL REFRACTARIA EN PACIENTES CON ENFERMEDAD DE CROHN ENSAYO CLNICO DE FASE II ANTECEDENTES:Las fístulas perianales de la enfermedad de Crohn a menudo son resistentes a las terapias anti-TNF-α estándares. Los exosomas derivados de células madre mesenquimales (MSC) son vesículas extracelulares que tienen efectos antiinflamatorios muy potentes, y la fase anterior de este estudio demostró su seguridad en el tratamiento de fístulas perianales refractarias.OBJETIVO:Evaluar la eficacia de los exosomas derivados de MSC para el tratamiento de fístulas perianales refractarias.DISEÑO:Ensayo clínico de fase II, no aleatorizado y no ciego, unicéntrico.LUGARES:Hospital universitario terciario.PACIENTES:Se inscribieron veintitrés pacientes, 20 de los cuales completaron el estudio. La fístula perianal refractaria se definió como la resistencia a al menos un ciclo de tratamiento con terapia anti-TNF-α.INTERVENCIONES:Después de la evaluación clínica y la resonancia magnética, los pacientes fueron sometidos a anestesia general y se inyectaron 5 ml de solución de exosoma directamente en los trayectos de la fístula. Las inyecciones se repitieron tres veces a intervalos de 2 meses y los pacientes fueron seguidos mensualmente durante 6 meses después de la última inyección. Se obtuvieron muestras de tejido de los tractos antes de cada inyección y se sometieron a evaluación inmunohistopatológica. Los datos de imágenes de resonancia magnética se obtuvieron antes y seis meses después de la última inyección.PRINCIPALES MEDIDAS DE RESULTADO:El resultado primario de este estudio fue el cierre del trayecto de la fístula en el examen clínico y la imagen de resonancia magnética. El resultado secundario fue una mejora en la descarga de los tractos.RESULTADOS:Los trayectos de la fístula se cerraron completamente en 12 (60%) de los pacientes. Cuatro pacientes mostraron mejoría clínica, algunos tractos permanecieron abiertos y cuatro pacientes fueron completamente resistentes al tratamiento. Durante el ensayo se trataron un total de 43 trayectos fistulosos, 30 (69,7%) de los cuales mostraron un cierre completo. El análisis histopatológico reveló reducciones sustanciales en la inflamación local y signos de una mayor regeneración tisular. El análisis inmunohistoquímico del grupo de diferenciación 68, 20 y 31 reafirmó estos resultados.CONCLUSIONES:Los exosomas derivados de MSC son seguros y eficaces para el tratamiento de fístulas perianales refractarias en pacientes con enfermedad de Crohn. (Traducción-Dr. Aurian Garcia Gonzalez ).
Collapse
Affiliation(s)
- Alireza Hadizadeh
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Female Pelvic Medicine and Reconstructive Surgery (FPMRS) Division, University of Chicago Pritzker School of Medicine, Northshore University HealthSystem, Skokie, Illinois
| | - Reza Akbari Asbagh
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Asieh Heirani-Tabasi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parastou Gorovanchi
- Department of Pathology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Ebrahimi Daryani
- Department of Gastroenterology, Division of Gastroenterology, Imam Khomeini Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Vahedi
- Department of Pathology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hengameh Nazari
- Radiology Department, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyedeh-Parnian Banikarimi
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahnoosh Abbaszade Dibavar
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Behboudi
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadegh Fazeli
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Keramati
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Keshvari
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Kazemeini
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Pak
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Reza Fazeli
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Foroogh Alborzi Avanaki
- Department of Gastroenterology, Division of Gastroenterology, Imam Khomeini Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed-Mohsen Ahmadi-Tafti
- Colorectal Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Yun JH, Lee HY, Yeou SH, Jang JY, Kim CH, Shin YS, D'Lima DD. Electrostatic attachment of exosome onto a 3D-fabricated calcium silicate/polycaprolactone for enhanced bone regeneration. Mater Today Bio 2024; 29:101283. [PMID: 39415763 PMCID: PMC11480244 DOI: 10.1016/j.mtbio.2024.101283] [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: 07/21/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Exosomes have garnered attention for use in bone regeneration, but their low activity, rapid degradation, and inaccurate delivery have been obstacles to their use in clinical applications. As such, there exists a need for an exosome-integrated delivery platform. Calcium silicate (Ca-Si) is considered one of the most promising bioceramics for bone regeneration because of its remarkable ability to promote hydroxyapatite formation, osteoblast proliferation, and differentiation. However, Ca-Si has limitations, such as a high degradation rate leading to high pH values. Here, we propose a bone regeneration platform: three-dimensional-fabricated Ca-Si scaffolds immersed in polycaprolactone (PCL) coated with exosomes. This setup enhanced porosity, mechanical strength, and natural hydroxyapatite formation. Ca-Si incorporation increased the quantity of attached exosomes on the scaffold and enabled more sustainable control of their release compared to bare PCL. The exosome-coated scaffold exhibited excellent cell attachment and osteogenic differentiation, significantly increasing biocompatibility and the in situ recruitment of stem cells when transplanted into the subcutaneous tissue of mice. The bone regenerating efficacy of the exosome-attached scaffold was confirmed using a mouse calvarial bone defect animal model. These findings suggest a potential application of exosome-coated Ca-Si/PCL scaffolds as an osteogenic platform for critical bone defects.
Collapse
Affiliation(s)
- Ju Hyun Yun
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Ewha Womans University, Seoul, 07985, Republic of Korea
| | - Hye-Young Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Se Hyun Yeou
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
- Scripps Health, Shiley Center for Orthopedic Research and Education at Scripps Clinic, La Jolla, CA, 92121, USA
| | - Darryl D. D'Lima
- Scripps Health, Shiley Center for Orthopedic Research and Education at Scripps Clinic, La Jolla, CA, 92121, USA
| |
Collapse
|
31
|
Zhang K, Wu D, Huang C. Crosstalk between non-coding RNA and apoptotic signaling in diabetic nephropathy. Biochem Pharmacol 2024; 230:116621. [PMID: 39542182 DOI: 10.1016/j.bcp.2024.116621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/18/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Diabetic nephropathy (DN) is a leading cause of end-stage renal disease in diabetes mellitus. It is also a significant contributor to cardiovascular morbidity and mortality in diabetic patients Thereby, Innovative therapeutic approaches are needed to retard the initiation and advancement of DN. Hyperglycemia can induce apoptosis, a regulated form of cell death, in multiple renal cell types, such as podocytes, mesangial cells, and proximal tubule epithelial cells, ultimately contributing to the pathogenesis of DN. Recent genome-wide investigations have revealed the widespread transcription of the human genome, resulting in the production of numerous regulatory non-protein-coding RNAs (ncRNAs), including microRNAs (miRNAs) and diverse categories of long non-coding RNAs (lncRNAs). They play a critical role in preserving physiological homeostasis, while their dysregulation has been implicated in a broad spectrum of disorders, including DN. Considering the established association between apoptotic processes and the expression of ncRNAs in DN, a thorough understanding of their intricate interplay is essential. Therefore, the current work thoroughly analyzes the intricate interplay among miRNAs, lncRNAs, and circular RNAs in the context of apoptosis within the pathogenesis of DN. Additionally, in the final section, we demonstrated that ncRNA-mediated modulation of apoptosis can be achieved through stem cell-derived exosomes and herbal medicines, presenting potential avenues for the treatment of DN.
Collapse
Affiliation(s)
- Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
32
|
Lin X, Fang Y, Mi X, Fu J, Chen S, Wu M, Jin N. Intrauterine injection of bioengineered hydrogel loaded exosomes derived from HUCM stem cells and spermidine prominently augments the pregnancy rate in thin endometrium rats. Regen Ther 2024; 27:63-72. [PMID: 38525237 PMCID: PMC10959642 DOI: 10.1016/j.reth.2024.02.003] [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: 11/30/2023] [Revised: 01/10/2024] [Accepted: 02/16/2024] [Indexed: 03/26/2024] Open
Abstract
The endometrium is essential to the development of embryos and pregnancy. Human umbilical cord mesenchymal stem cells (HUCMSCs) are promising stem cell sources. HUCMSCs self-renew quickly and are painless to collect. Spermidine is an inherent polyamine needed for cellular and molecular processes that regulate physiology and function. HUCMSCs and spermidine (SN) may heal intrauterine adhesions. HUCMSCs were investigated for endometrial repair in rats. Composite hydrogels are used for medical exosome implantation, including their materials, properties, and embedding procedures. This study examined whether bioengineered hydrogel-loaded exosomes from HUCMSCs and spermidine prenatally improved conception rates in mice with poor endometrial lining. The data show that HUCMSC and SN provide a good experimental base for HUCMSC safety and intrauterine treatment in rats. Western blots, exosome structural analysis, pregnancy outcomes, flow cytometry, H&E staining, immunohistochemistry, and immunofluorescence labelling found and recovered the aberrant area. HUCM-derived stem cells and spermidine-derived exosomes biophysically match. These traits strengthen and prolong endometrial function. Pregnant rats with HUCMSC and SN had thinner endometrium. Hydrogel-incorporated HEHUCMSC and SN exosomes may improve IUI in rats with thin endometrium.
Collapse
Affiliation(s)
- Xiuying Lin
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Yanbian University, Yanbian 133002, China
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Yanqiu Fang
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Xuguang Mi
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Jianhua Fu
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Shiling Chen
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Mengxue Wu
- Center of Reproductive Medicine, Jilin Province People's Hospital, Changchun 130021, China
| | - Ningyi Jin
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Yanbian University, Yanbian 133002, China
- Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun 130000, China
| |
Collapse
|
33
|
Zhou W, Jiang X, Gao J. Extracellular vesicles for delivering therapeutic agents in ischemia/reperfusion injury. Asian J Pharm Sci 2024; 19:100965. [PMID: 39640057 PMCID: PMC11617990 DOI: 10.1016/j.ajps.2024.100965] [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: 05/07/2024] [Revised: 06/08/2024] [Accepted: 06/29/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemia/reperfusion (I/R) injury is marked by the restriction and subsequent restoration of blood supply to an organ. This process can exacerbate the initial tissue damage, leading to further disorders, disability, and even death. Extracellular vesicles (EVs) are crucial in cell communication by releasing cargo that regulates the physiological state of recipient cells. The development of EVs presents a novel avenue for delivering therapeutic agents in I/R therapy. The therapeutic potential of EVs derived from stem cells, endothelial cells, and plasma in I/R injury has been actively investigated. Therefore, this review aims to provide an overview of the pathological process of I/R injury and the biophysical properties of EVs. We noted that EVs serve as nontoxic, flexible, and multifunctional carriers for delivering therapeutic agents capable of intervening in I/R injury progression. The therapeutic efficacy of EVs can be enhanced through various engineering strategies. Improving the tropism of EVs via surface modification and modulating their contents via preconditioning are widely investigated in preclinical studies. Finally, we summarize the challenges in the production and delivery of EV-based therapy in I/R injury and discuss how it can advance. This review will encourage further exploration in developing efficient EV-based delivery systems for I/R treatment.
Collapse
Affiliation(s)
- Weihang Zhou
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
34
|
Sankaranarayanan J, Lee SC, Kim HK, Kang JY, Kuppa SS, Seon JK. Exosomes Reshape the Osteoarthritic Defect: Emerging Potential in Regenerative Medicine-A Review. Int J Stem Cells 2024; 17:381-396. [PMID: 38246659 PMCID: PMC11612219 DOI: 10.15283/ijsc23108] [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: 07/08/2023] [Revised: 10/05/2023] [Accepted: 11/28/2023] [Indexed: 01/23/2024] Open
Abstract
Osteoarthritis (OA) is a joint disorder caused by wear and tear of the cartilage that cushions the joints. It is a progressive condition that can cause significant pain and disability. Currently, there is no cure for OA, though there are treatments available to manage symptoms and slow the progression of the disease. A chondral defect is a common and devastating lesion that is challenging to treat due to its avascular and aneural nature. However, there are conventional therapies available, ranging from microfracture to cell-based therapy. Anyhow, its efficiency in cartilage defects is limited due to unclear cell viability. Exosomes have emerged as a potent therapeutic tool for chondral defects because they are a complicated complex containing cargo of proteins, DNA, and RNA as well as the ability to target cells due to their phospholipidic composition and the altering exosomal contents that boost regeneration potential. Exosomes are used in a variety of applications, including tissue healing and anti-inflammatory therapy. As in recent years, biomaterials-based bio fabrication has gained popularity among the many printable polymer-based hydrogels, tissue-specific decellularized extracellular matrix might boost the effects rather than an extracellular matrix imitating environment, a short note has been discussed. Exosomes are believed to be the greatest alternative option for current cell-based therapy, and future progress in exosome-based therapy could have a greater influence in the field of orthopaedics. The review focuses extensively on the insights of exosome use and scientific breakthroughs centered OA.
Collapse
Affiliation(s)
- Jaishree Sankaranarayanan
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, Korea
- Department of Orthopaedic Surgery, Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Seok Cheol Lee
- Department of Orthopaedic Surgery, Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Hyung Keun Kim
- Department of Orthopaedic Surgery, Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Ju Yeon Kang
- Department of Orthopaedic Surgery, Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Sree Samanvitha Kuppa
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, Korea
- Department of Orthopaedic Surgery, Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Jong Keun Seon
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, Korea
- Department of Orthopaedic Surgery, Center for Joint Disease, Chonnam National University Hwasun Hospital, Hwasun, Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
35
|
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
|
36
|
Jaffet J, Singh V, Schrader S, Mertsch S. The Potential Role of Exosomes in Ocular Surface and Lacrimal Gland Regeneration. Curr Eye Res 2024:1-14. [PMID: 39508276 DOI: 10.1080/02713683.2024.2424265] [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: 08/01/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE Dry eye disease (DED), a multifactorial disease of the lacrimal system, manifests itself in patients with various symptoms such as itching, inflammation, discomfort and visual impairment. In its most severe forms, it results in the breakdown of the vital tissues of lacrimal functional unit and carries the risk of vision loss. Despite the frequency of occurrence of the disease, there are no effective curative treatment options available to date. Treatment using stem cells and its secreted factors could be a promising approach in the regeneration of damaged tissues of ocular surface. The treatment using secreted factors as well as extracellular vesicles has been demonstrated beneficial effects in various ocular surface diseases. This review provides insights on the usage of stem cell derived exosomes as a promising therapy against LG dysfunction induced ADDE for ocular surface repair. METHODS In order to gain an overview of the existing research in this field, literature search was carried out using the PubMed, Medline, Scopus and Web of Science databases. This review is based on 164 publications until June 2024 and the literature search was carried out using the key words "exosomes", "lacrimal gland regeneration", "exosomes in lacrimal dysfunction". RESULTS The literature and studies till date suggest that exosomes and other secreted factors from stem cells have demonstrated beneficial effects on damaged ocular tissues in various ocular surface diseases. Exosomal cargo plays a crucial role in regenerating tissues by promoting homeostasis in the lacrimal system, which is often compromised in severe cases of dry eye disease. Exosome therapy shows promise as a regenerative therapy, potentially addressing the lack of effective curative treatments available for patients with dry eye disease. CONCLUSION Stem cell-derived exosomes represent a promising, innovative approach as a new treatment option for ADDE. By targeting lacrimal gland dysfunction and enhancing ocular surface repair, exosome therapy offers potential for significant advances in dry eye disease management. Future research is needed to refine the application of this therapy, optimize delivery methods, and fully understand its long-term efficacy in restoring ocular health.
Collapse
Affiliation(s)
- Jilu Jaffet
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
- LV Prasad Eye Institute, Centre for Ocular Regeneration, Hyderabad, Telangana, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Vivek Singh
- LV Prasad Eye Institute, Centre for Ocular Regeneration, Hyderabad, Telangana, India
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
| |
Collapse
|
37
|
Košuthová H, Fecskeová LK, Matejová J, Slovinská L, Morávek M, Bártová Z, Harvanová D. Effects of Replicative Senescence of Human Chorionic MSCs on their EV-miRNA Profile. Stem Cell Rev Rep 2024; 20:2318-2335. [PMID: 39305404 PMCID: PMC11554840 DOI: 10.1007/s12015-024-10790-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2024] [Indexed: 11/12/2024]
Abstract
Chorionic mesenchymal stromal cells (CHO-MSCs) and their extracellular vesicles (EVs) are becoming increasingly popular, since chorion is ethically harmless and an easily accessible source of MSCs. However, until now there is only a limited number of studies with a thorough characterization of CHO-MSCs derived EVs and their miRNA profile. In this study, we monitored changes in the EV-miRNA profile between early and late passage of human CHO-MSCs. First, senescence of CHO-MSCs was induced by serial passaging and confirmed by morphological changes, shortened telomeres and changes in the expression of selected genes. The expression of MSCs-specific surface markers CD73, CD90, CD105 did not change with increasing passages. Next, EVs and their miRNA profiles were compared between early vs late passage cells. Number of EVs and their size were not significantly changed. Seven of the top 10 most expressed EV-miRNAs were common to both early and late passages. A differential expression study between early and late passages identified 37 significantly differentially expressed EV-miRNAs, out of which 23 were found to be associated with pathways of cellular senescence based on KEGG pathway analysis. A set of 9 miRNAs were identified as the most frequently associated with senescence and/or with the most altered expression between early and late passages, out of which miR-145-5p, miR-335-5p and miR-199b-3p were the most significant downregulated miRNAs in late passages. The most upregulated EV-miRNAs were miR-1307-3p, miR-3615 and miR320b. Targeting these miRNAs in future experiments may prolong the therapeutic potential of CHO-MSCs and their EVs.
Collapse
Affiliation(s)
- Hedviga Košuthová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Lívia K Fecskeová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia.
| | - Jana Matejová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Lucia Slovinská
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Marko Morávek
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| | - Zuzana Bártová
- Institute of Geotechnics of the Slovak Academy of Sciences, Watsonova 45, 040 01, Kosice, Slovakia
| | - Denisa Harvanová
- Associated Tissue Bank, Faculty of Medicine, Pavol Jozef Safarik University and Louis Pasteur University Hospital, Trieda SNP 1, 04011, Kosice, Slovakia
| |
Collapse
|
38
|
Altug B, Soykan MN, Eyubova S, Eker Sariboyaci A, Dogan C, Ozalp O, Atalay E. Crosstalk among miR-29, α-SMA, and TGFβ1/β3 in melatonin-induced exosome (Mel-prExo) treated human limbal mesenchymal stem cells (hLMSCs): An insight into scarless healing of the cornea. Biofactors 2024; 50:1287-1297. [PMID: 38804543 PMCID: PMC11627467 DOI: 10.1002/biof.2085] [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: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Inflammatory mediators that infiltrate the corneal stroma after corneal infections, trauma or refractive surgery can trigger the transformation of corneal keratocytes into myofibroblasts, resulting in highly irregular collagen deposition and subsequently corneal scarring. Mesenchymal stem cells (MSCs) can be used as therapeutic agents to regenerate corneal and conjunctival tissue damage, regulate inflammation, and reduce the development of limbal stem cell failure. The use of MSC-derived exosomes as a cell-free therapeutic vector is a novel therapeutic approach. This study aimed to assess the effect of exosomes obtained from melatonin (Mel)-treated human limbal mesenchymal stem cells (hLMSCs) on naïve hLMSCs and to determine their influence on the antifibrotic and pro-regenerative pathways involved in corneal scarring. hLMSCs were treated with varying concentrations of Mel, followed by isolation and characterization of the procured exosomes (Mel-prExos). These exosomes were added to the cell culture media of naïve hLMSCs to examine their antifibrotic and pro-regenerative effects. The expression of miR-155, miR-29, TGFβ1, TGFβ3, PPARγ, and α-SMA miRNAs and genes were compared between Mel-treated hLMSCs and Mel-prExo-treated hLMSCs by using real-time PCR. We found that at 1 μM Mel and in the presence of Mel-prExos, TGFβ1 was expressed 0.001-fold, while TGFβ3 was expressed 0.6-fold. miR-29 expression was increased 38-fold in the control-Exo group compared to that in the control group. Changes in TGFβ1/β3 and α-SMA expression are associated with miR-29 and miR-155. This approach could prove beneficial for ocular surface tissue engineering applications.
Collapse
Affiliation(s)
- Burcugul Altug
- Cellular Therapy and Stem Cell Production Application, Research Centre (ESTEM)Eskisehir Osmangazi UniversityEskisehirTürkiye
| | - Merve Nur Soykan
- Cellular Therapy and Stem Cell Production Application, Research Centre (ESTEM)Eskisehir Osmangazi UniversityEskisehirTürkiye
- Department of Stem CellInstitute of Health Sciences, Eskisehir Osmangazi UniversityEskisehirTürkiye
| | - Sevinc Eyubova
- Cellular Therapy and Stem Cell Production Application, Research Centre (ESTEM)Eskisehir Osmangazi UniversityEskisehirTürkiye
- Department of Stem CellInstitute of Health Sciences, Eskisehir Osmangazi UniversityEskisehirTürkiye
| | - Ayla Eker Sariboyaci
- Cellular Therapy and Stem Cell Production Application, Research Centre (ESTEM)Eskisehir Osmangazi UniversityEskisehirTürkiye
- Department of Stem CellInstitute of Health Sciences, Eskisehir Osmangazi UniversityEskisehirTürkiye
| | - Cezmi Dogan
- Department of Ophthalmologyİstanbul University‐Cerrahpaşa, Cerrahpaşa Faculty of MedicineİstanbulTürkiye
| | - Onur Ozalp
- Department of Ophthalmology, Faculty of MedicineEskisehir Osmangazi UniversityEskisehirTürkiye
| | - Eray Atalay
- Cellular Therapy and Stem Cell Production Application, Research Centre (ESTEM)Eskisehir Osmangazi UniversityEskisehirTürkiye
- Department of Ophthalmology, Faculty of MedicineEskisehir Osmangazi UniversityEskisehirTürkiye
| |
Collapse
|
39
|
Kim M, Kim TH, Salas ESS, Jeon S, Shin JH, Choi D. The efficacy of exosomes from human chemically derived hepatic progenitors in liver damage alleviation: a preclinical experimental study. Ann Surg Treat Res 2024; 107:252-263. [PMID: 39524547 PMCID: PMC11543897 DOI: 10.4174/astr.2024.107.5.252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/22/2024] [Accepted: 08/18/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose Over the past decade, interest in exosomes as therapeutics has surged. In particular, stem-cell-derived exosomes may be more effective as a treatment for liver disease than the stem cells themselves. We have previously developed human chemically derived hepatic progenitors (hCdHs) from human hepatocytes. hCdHs can differentiate into hepatocytes and cholangiocytes, regenerating the liver in mouse models. In this study, we evaluated the mitigating effects of hCdHs-derived exosomes (hCdHs-exo) on liver damage and compared them with those of exosomes from bone marrow mesenchymal stem cells (BMMSCs-exo). Methods Exosomes were isolated from hCdHs and BMMSCs by culturing cells in large quantities and separating the exosomes from the culture medium using ultracentrifugation. Isolated exosomes were characterized by various methods before experimental use. In vitro, the ability of exosomes to inhibit activation of hepatic stellate cells (HSCs) by transforming growth factor beta 1 was evaluated. In vivo, exosomes were injected into mice with carbon tetrachloride (CCl4)-induced liver damage, and their effectiveness in mitigating liver damage was assessed by histological staining and biochemical analysis. Results The analyses confirmed the successful isolation of exosomes from both cell types. In vitro, hCdHs-exo significantly reduced the levels of transcription factors and activation markers in induced HSCs. In vivo, hCdHs-exo effectively alleviated liver damage caused by CCl4. Furthermore, both in vitro and in vivo studies confirmed that hCdHs-exo had a greater effect in alleviating liver damage than did BMMSCs-exo. Conclusion These results demonstrate that hCdHs-exo, similarly to hCdHs, have superior efficacy in alleviating liver damage compared with BMMSCs-exo.
Collapse
Affiliation(s)
- Min Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Tae Hun Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Elsy Soraya Silva Salas
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Soyoung Jeon
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Ji Hyun Shin
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| |
Collapse
|
40
|
Motlagh Asghari K, Novinbahador T, Mehdizadeh A, Zolfaghari M, Yousefi M. Revolutionized attitude toward recurrent pregnancy loss and recurrent implantation failure based on precision regenerative medicine. Heliyon 2024; 10:e39584. [PMID: 39498089 PMCID: PMC11532865 DOI: 10.1016/j.heliyon.2024.e39584] [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: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Traditional treatment strategies for recurrent pregnancy loss (RPL) and recurrent implantation failure (RIF) often result in limited success, placing significant emotional and financial burdens on couples. However, novel approaches such as diagnostic gene profiling, cell therapy, stem cell-derived exosome therapy, and pharmacogenomics offer promising, personalized treatments. Combining traditional treatments with precision and regenerative medicine may enhance the efficacy of these approaches and improve pregnancy outcomes. This review explores how integrating these strategies can potentially transform the lives of couples experiencing repeated pregnancy loss or implantation failure, providing hope for improved treatment success. Precision and regenerative medicine represent a new frontier for managing RPL and RIF, offering promising solutions.
Collapse
Affiliation(s)
| | - Tannaz Novinbahador
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Yin B, Xu J, Lu J, Ou C, Zhang K, Gao F, Zhang Y. Responsive Hydrogel-Based Drug Delivery Platform for Osteoarthritis Treatment. Gels 2024; 10:696. [PMID: 39590052 PMCID: PMC11594092 DOI: 10.3390/gels10110696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disorder and is a major cause of disability among the elderly population. The degeneration and damage of articular cartilage associated with OA can result in a diminished range of motion in joints, subsequently impacting fundamental activities such as ambulation, standing, and grasping objects. In severe cases, it may culminate in disability. Traditional pharmacological treatments are often accompanied by various side effects, while invasive surgical procedures increase the risk of infection and thrombosis. Consequently, identifying alternative new methods for OA treatment remains a formidable challenge. With advancements in responsive hydrogel drug delivery platforms, an increasing number of strategies have emerged to enhance OA treatment protocols. Injectable response hydrogel drug delivery platforms show many advantages in treating OA, including improved biocompatibility, prolonged drug release duration, elevated drug loading capacity and enhanced sensitivity. This article reviews the recent progress of injectable responsive hydrogel drug delivery platform for OA treatment over the past few years. These innovative methodologies present new strategies and directions for future OA treatment while summarizing a series of challenges faced during the clinical transformation of injectable response hydrogel drug delivery platforms. Overall, injectable responsive hydrogel drug delivery platforms show great potential in treating OA, especially regarding improving drug retention time and stimulus-responsive release at the lesion sites. These innovative methods provide new hope for future OA treatment and point the way for clinical applications.
Collapse
Affiliation(s)
- Bin Yin
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, China; (B.Y.); (J.L.); (K.Z.); (F.G.)
| | - Jianda Xu
- Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou 213003, China;
| | - Jingqi Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, China; (B.Y.); (J.L.); (K.Z.); (F.G.)
| | - Changjin Ou
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, China; (B.Y.); (J.L.); (K.Z.); (F.G.)
| | - Kai Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, China; (B.Y.); (J.L.); (K.Z.); (F.G.)
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, China; (B.Y.); (J.L.); (K.Z.); (F.G.)
| | - Yizhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing 210044, China; (B.Y.); (J.L.); (K.Z.); (F.G.)
| |
Collapse
|
42
|
Zhang Y, Li D, Han Y, Wu M, Zhang S, Ma H, Liu L, Ju X. Intraovarian injection of 3D-MSC-EVs-ECM gel significantly improved rat ovarian function after chemotherapy. Reprod Biol Endocrinol 2024; 22:125. [PMID: 39415205 PMCID: PMC11481453 DOI: 10.1186/s12958-024-01299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Restoring the function of the ovary is important for chemotherapy-induced ovarian failure (COF) patients. Stem cell and extracellular vesicles (EVs) therapy show promise but need further improvement. METHODS Human umbilical cord mesenchymal stem cells (hUC-MSCs) were primarily cultured and further three-dimensional (3D) cultured using an ultra-low attachment surface method. The expression levels of nutritional cytokines and immunomodulatory and stemness-related genes of 3D-cultured hUC-MSCs were analyzed. EVs were isolated by ultracentrifugation and characterized. Ovaries were decellularized with sodium dodecyl sulfate to obtain extracellular matrix (ECM). Lyophilized EVs from three-dimensional (2D) or 3D hUC-MSCs were mixed with ECM to prepare the 2D/3D-MSC-EVs-ECM gels. The therapeutic effect of the MSC-EVs-ECM gel on cyclophosphamide (CTX) -treated rats was analyzed through various tests. RNA sequencing was used to analyze the expression changes of genes before and after treatment. RESULTS After culturing in ultra-low attachment dishes, hUC-MSCs aggregated into spheroids and significantly upregulated the expression levels of immunomodulatory and stemness-related genes. The total EVs yield was also upregulated (5.6-fold) after 3D culture. The cell viability of CTX-treated ovarian granulosa cells (OGCs) was significantly rescued by coculture with the 3D-MSC-EVs-ECM gel. Hormones indicative of ovarian function, AMH, E2, and FSH, were recovered in both the CTX + 2D-MSC-EVs-ECM gel group and the CTX + 3D-MSC-EVs-ECM gel group, while the apoptosis-related protein Bax was significantly downregulated. The 3D-MSC-EVs-ECM gel was more effective than the 2D-MSC-EVs-ECM gel. Significantly differentially expressed genes, such as Hbb-b1, Gpd1, and Sirpa, were detected by RNA sequencing. Hbb-b1 was increased in the ovaries of CTX-treated rats, and this increase was attenuated by injecting the 2D/3D-MSC-EVs-ECM gel. Gpd1 was increased after CTX treatment, and this increase was reversed by the 3D-MSC-EVs-ECM gel. Sirpa was decreased in the ovaries of CTX-treated rats, and this decrease was attenuated by injecting the 3D-MSC-EVs-ECM gel. CONCLUSIONS Our study demonstrated that the 3D-MSC-EVs-ECM gel is an efficient strategy for the recovery of ovarian function in CTX-induced ovarian failure.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Department of Anesthesiology, Shanghai Jiaotong University First People's Hospital (Shanghai General Hospital), Shanghai, China
| | - Dong Li
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Yi Han
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Min Wu
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Shule Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Huixian Ma
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Linghong Liu
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China.
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
43
|
Sani F, Shafiei F, Dehghani F, Mohammadi Y, Khorraminejad‐Shirazi M, Anvari‐Yazdi AF, Moayedfard Z, Azarpira N, Sani M. Unveiling exosomes: Cutting-edge isolation techniques and their therapeutic potential. J Cell Mol Med 2024; 28:e70139. [PMID: 39431552 PMCID: PMC11492151 DOI: 10.1111/jcmm.70139] [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: 06/16/2024] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024] Open
Abstract
Exosomes are one type of nanosized membrane vesicles with an endocytic origin. They are secreted by almost all cell types and play diverse functional roles. It is essential for research purposes to differentiate exosomes from microvesicles and isolate them from other components in a fluid sample or cell culture medium. Exosomes are important mediators in cell-cell communication. They deliver their cargos, such as mRNA transcripts, microRNA, lipids, cytosolic and membrane proteins and enzymes, to target cells with or without physical connections between cells. They are highly heterogeneous in size, and their biological functions can vary depending on the cell type, their ability to interact with recipient cells and transport their contents, and the environment in which they are produced. This review summarized the recent progress in exosome isolation and characterization techniques. Moreover, we review the therapeutic approaches, biological functions of exosomes in disease progression, tumour metastasis regulation, immune regulation and some ongoing clinical trials.
Collapse
Affiliation(s)
- Farnaz Sani
- Shiraz Institute for Stem Cell & Regenerative MedicineShiraz University of Medical SciencesShirazIran
| | - Faezeh Shafiei
- Shiraz Institute for Stem Cell & Regenerative MedicineShiraz University of Medical SciencesShirazIran
| | - Farshad Dehghani
- Shiraz Institute for Stem Cell & Regenerative MedicineShiraz University of Medical SciencesShirazIran
| | - Yasaman Mohammadi
- Pharmaceutical Sciences Research CenterShiraz University of Medical ScienceShirazIran
| | - Mohammadhossein Khorraminejad‐Shirazi
- Department of Pathology, School of MedicineShiraz University of Medical SciencesShirazIran
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
- Department of Pathology, School of MedicineJahrom University of Medical SciencesJahromIran
| | | | - Zahra Moayedfard
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Negar Azarpira
- Shiraz Institute for Stem Cell & Regenerative MedicineShiraz University of Medical SciencesShirazIran
- Transplant Research CenterShiraz University of Medical SciencesShirazIran
| | - Mahsa Sani
- Shiraz Institute for Stem Cell & Regenerative MedicineShiraz University of Medical SciencesShirazIran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| |
Collapse
|
44
|
Puspita R, Jusuf AA, Antarianto RD, Sianipar IR. A systematic review of the anti-inflammatory and anti-fibrotic potential of human umbilical cord mesenchymal stem cells-derived exosomes in experimental models of liver regeneration. Mol Biol Rep 2024; 51:999. [PMID: 39302506 DOI: 10.1007/s11033-024-09929-0] [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: 07/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Chronic liver injuries and their complications are leading causes of death, especially in developing countries (Sharma and Nagalli in Sex/Gender-Specific Medicine in the Gastrointestinal Diseases, StatPearls Publishing, 2023). The available and effective treatment plans are limited, implicating the need for innovative treatment approaches (Tsuchiya et al. in Inflamm Regener, 2019;Sharma and Nagalli in Sex/Gender-Specific Medicine in the Gastrointestinal Diseases, StatPearls Publishing, 2023;Younossi et al. in Clin Gastroenterol Hepatol 21:1978-1991, 2023;). This paper aims to summarize the effects and mechanisms of hUC-MSC-exo on liver injuries and its complications; it also suggests future directions for future research. The outcomes of interest are the morphology and histology of the liver, pathology score, liver function enzyme, glucose and lipid metabolism, and the effect hUC-MSC-exo had on gene regulation regarding liver diseases. A comprehensive review of nineteen studies was conducted to assess the effectiveness of the implementation of the hUC-MSC-Exo, instilling confidence in the validity of the findings. Regarding the morphology and histology of the liver and pathology score, hUC-MSC-exo treatment resulted in improved liver morphology post-treatment, as indicated by the reduction in pathology scores. However, these observed improvements in the liver surface are not directly attributed to the hUC-MSC-Exo itself but to the overall healing processes stimulated by the treatment. In physiological outcomes, hUC-MSC-exo also improves glucose and lipid metabolism, especially in diet-induced liver injury and its complications. In gene regulation, one interesting gene in this intervention is the fat mass and obesity-associated (FTO), in which hUC-MSC-exo combined with miRNAs can suppress FTO. HUC-MSC-Exo can improve by utilizing several possible pathways, targeting pinpoints in the pathogenesis of liver disease or glucose and lipid metabolism. This study presents hUC-MSC-exo better in all outcomes of interest compared to the control or sham group. Further specification of indications of the hUC-MSC-exo method may be beneficial and essential to be analyzed in future reviews to better understand the effectiveness of each hUC-MSC-exo dose, duration, and medium.
Collapse
Affiliation(s)
- Ratna Puspita
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Biochemistry, Faculty of Medicine, Universitas Pembangunan Nasional Veteran Jakarta, Jakarta, Indonesia
| | - Ahmad Aulia Jusuf
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
| | | | | |
Collapse
|
45
|
Jabermoradi S, Paridari P, Ramawad HA, Gharin P, Roshdi S, Toloui A, Yousefifard M. Stem Cell-Derived Exosomes as a Therapeutic Option for Spinal Cord Injuries; a Systematic Review and Meta-Analysis. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2024; 13:e2. [PMID: 39318865 PMCID: PMC11417640 DOI: 10.22037/aaem.v12i1.2261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Introduction Exosomes function as cell signaling carriers and have drawn much attention to the cell-free treatments of regenerative medicine. This meta-analysis aimed to investigate the efficacy of mesenchymal stem cell-derived (MSC-derived) exosomes in animal models of spinal cord injuries (SCI). Method A comprehensive search was conducted in Medline, Embase, Scopus, and Web of Science to attain related articles published by January 31, 2023. The eligible keywords were correlated with the spinal cord injury and MSC-derived exosomes. The evaluated outcomes were locomotion, cavity size, cell apoptosis, inflammation, neuro-regeneration, and microglia activation. A standardized mean difference was calculated for each sample and a pooled effect size was reported. Results 65 papers fully met the inclusion criteria. Treatment with MSC-derived exosomes ultimately improved locomotion and shrunk cavity size (p<0.0001). The administration of MSC-derived exosomes enhanced the expression of beta-tubulin III, NF200, and GAP-43, and increased the number of NeuN-positive and Nissl-positive cells, while reducing the expression of glial fibrillary acidic protein (p<0.0001). The number of apoptotic cells in the treatment group decreased significantly (p<0.0001). Regarding the markers of microglia activation, MSC-derived exosomes increased the number of CD206- and CD68-positive cells (p=0.032 and p<0.0001, respectively). Additionally, MSC-derived exosome administration significantly increased the expression of the anti-inflammatory interleukin (IL)-10 and IL-4 (p<0.001 and p=0.001, respectively) and decreased the expression of the inflammatory IL-1b, IL-6, and TNF-a (p<0.0001). Conclusion MSC-derived exosome treatment resulted in a significantly improved locomotion of SCI animals through ameliorating neuroinflammation, reducing apoptosis, and inducing neuronal regrowth by facilitating a desirable microenvironment.
Collapse
Affiliation(s)
- Sajjad Jabermoradi
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
- The first and second authors have identical contributions
| | - Parsa Paridari
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
- The first and second authors have identical contributions
| | - Hamzah Adel Ramawad
- Department of EmergencyMedicine, NYC Health + Hospitals, Coney Island, New York, USA
| | - Pantea Gharin
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| | - Shayan Roshdi
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| | - Amirmohammad Toloui
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Montagnoli TL, Santos AD, Sudo SZ, Gubert F, Vasques JF, Mendez-Otero R, de Sá MPL, Zapata-Sudo G. Perspectives on Stem Cell Therapy in Diabetic Neuropathic Pain. Neurol Int 2024; 16:933-944. [PMID: 39311343 PMCID: PMC11417725 DOI: 10.3390/neurolint16050070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Diabetes mellitus-related morbidity and mortality are primarily caused by long-term complications such as retinopathy, nephropathy, cardiomyopathy, and neuropathy. Diabetic neuropathy (DN) involves the progressive degeneration of axons and nerve fibers due to chronic exposure to hyperglycemia. This metabolic disturbance leads to excessive activation of the glycolytic pathway, inducing oxidative stress and mitochondrial dysfunction, ultimately resulting in nerve damage. There is no specific treatment for painful DN, and new approaches should aim not only to relieve pain but also to prevent oxidative stress and reduce inflammation. Given that existing therapies for painful DN are not effective for diabetic patients, mesenchymal stromal cells (MSCs)-based therapy shows promise for providing immunomodulatory and paracrine regulatory functions. MSCs from various sources can improve neuronal dysfunction associated with DN. Transplantation of MSCs has led to a reduction in hyperalgesia and allodynia, along with the recovery of nerve function in diabetic rats. While the pathogenesis of diabetic neuropathic pain is complex, clinical trials have demonstrated the importance of MSCs in modulating the immune response in diabetic patients. MSCs reduce the levels of inflammatory factors and increase anti-inflammatory cytokines, thereby interfering with the progression of DM. Further investigation is necessary to ensure the safety and efficacy of MSCs in preventing or treating neuropathic pain in diabetic patients.
Collapse
Affiliation(s)
- Tadeu Lima Montagnoli
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (T.L.M.); (A.D.S.)
| | - Aimeé Diogenes Santos
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (T.L.M.); (A.D.S.)
| | - Susumu Zapata Sudo
- Programa de Pós-Graduação em Medicina (Cirurgia), Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.Z.S.); (M.P.L.d.S.)
| | - Fernanda Gubert
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Juliana Ferreira Vasques
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.F.V.); (R.M.-O.)
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.F.V.); (R.M.-O.)
| | - Mauro Paes Leme de Sá
- Programa de Pós-Graduação em Medicina (Cirurgia), Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.Z.S.); (M.P.L.d.S.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Gisele Zapata-Sudo
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (T.L.M.); (A.D.S.)
- Programa de Pós-Graduação em Medicina (Cirurgia), Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.Z.S.); (M.P.L.d.S.)
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
47
|
Akhlaghpasand M, Tavanaei R, Hosseinpoor M, Yazdani KO, Soleimani A, Zoshk MY, Soleimani M, Chamanara M, Ghorbani M, Deylami M, Zali A, Heidari R, Oraee-Yazdani S. Safety and potential effects of intrathecal injection of allogeneic human umbilical cord mesenchymal stem cell-derived exosomes in complete subacute spinal cord injury: a first-in-human, single-arm, open-label, phase I clinical trial. Stem Cell Res Ther 2024; 15:264. [PMID: 39183334 PMCID: PMC11346059 DOI: 10.1186/s13287-024-03868-0] [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: 02/01/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Neurological and functional impairments are commonly observed in individuals with spinal cord injury (SCI) due to insufficient regeneration of damaged axons. Exosomes play a crucial role in the paracrine effects of mesenchymal stem cells (MSCs) and have emerged as a promising therapeutic approach for SCI. Thus, this study aimed to evaluate the safety and potential effects of intrathecal administration of allogeneic exosomes derived from human umbilical cord MSCs (HUC-MSCs) in patients with complete subacute SCI. METHODS This study was a single-arm, open-label, phase I clinical trial with a 12-month follow-up period. HUC-MSCs were extracted from human umbilical cord tissue, and exosomes were isolated via ultracentrifugation. After intrathecal injection, each participant a underwent complete evaluation, including neurological assessment using the American Spinal Injury Association (ASIA) scale, functional assessment using the Spinal Cord Independence Measure (SCIM-III), neurogenic bowel dysfunction (NBD) assessment using the NBD score, modified Ashworth scale (MAS), and lower urinary tract function questionnaire. RESULTS Nine patients with complete subacute SCI were recruited. The intrathecal injection of allogeneic HUC-MSCs-exosomes was safe and well tolerated. No early or late adverse event (AE) attributable to the study intervention was observed. Significant improvements in ASIA pinprick (P-value = 0.039) and light touch (P-value = 0.038) scores, SCIM III total score (P-value = 0.027), and NBD score (P-value = 0.042) were also observed at 12-month after the injection compared with baseline. CONCLUSIONS This study demonstrated that intrathecal administration of allogeneic HUC-MSCs-exosomes is safe in patients with subacute SCI. Moreover, it seems that this therapy might be associated with potential clinical and functional improvements in these patients. In this regard, future larger phase II/III clinical trials with adequate power are highly required. TRIAL REGISTRATION Iranian Registry of Clinical Trials, IRCT20200502047277N1. Registered 2 October 2020, https://en.irct.ir/trial/48765 .
Collapse
Affiliation(s)
- Mohammadhosein Akhlaghpasand
- Medical Biotechnology Research Center, AJA University of Medical Sciences, PO box: 1411718541, Tehran, Iran
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Roozbeh Tavanaei
- Medical Biotechnology Research Center, AJA University of Medical Sciences, PO box: 1411718541, Tehran, Iran
| | - Maede Hosseinpoor
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
- Stem Cell Technology Research Center (STRC), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Kaveh Oraii Yazdani
- Department of cardiovascular diseases, Zahedan university of medical science, Zahedan, Iran
| | - Afsane Soleimani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Mojtaba Yousefi Zoshk
- Department of Pediatrics, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mahdi Ghorbani
- Medical Biotechnology Research Center, AJA University of Medical Sciences, PO box: 1411718541, Tehran, Iran
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Deylami
- Department of ICU &Critical care, Faculty of Medicine, Loghman-e Hakim Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran
| | - Reza Heidari
- Medical Biotechnology Research Center, AJA University of Medical Sciences, PO box: 1411718541, Tehran, Iran.
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran.
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, PO box: 1988873554, Tehran, Iran.
| |
Collapse
|
48
|
Fakouri A, Razavi ZS, Mohammed AT, Hussein AHA, Afkhami H, Hooshiar MH. Applications of mesenchymal stem cell-exosome components in wound infection healing: new insights. BURNS & TRAUMA 2024; 12:tkae021. [PMID: 39139205 PMCID: PMC11319788 DOI: 10.1093/burnst/tkae021] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/12/2024] [Accepted: 04/22/2024] [Indexed: 08/15/2024]
Abstract
The healing process at a wound is made up of many types of cells, growth factors, the extracellular matrix, nerves and blood vessels all interacting with each other in complex and changing ways. Microbial colonization and proliferation are possible at the place of injury, which makes infection more likely. Because of this, any cut has a chance of getting an infection. Researchers have found that wound infections make patients more upset and cost the healthcare system a lot of money. Surgical site infections happen a lot to people who have recently had surgery. This study shows that such surgical infection is linked to a high rate of illness and death. This is shown by the fact that 25% of patients get serious sepsis and need to be transferred to an intensive care unit. In both animal models and people, mesenchymal stem cells (MSCs) play an active role in all stages of wound healing and have positive effects. Exosomes are one of the main things MSCs release. They have effects that are similar to those of the parent MSCs. Various effector proteins, messenger RNA and microRNAs can be transported by extracellular vesicles to control the activity of target cells. This has a big impact on the healing process. These results suggest that using MSC-exosomes as a new type of cell-free therapy could be a better and safer option than whole cell therapy. This review is mostly about how to use parts of MSC-exosomes to help wound infections heal.
Collapse
Affiliation(s)
- Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad 6813833946, Iran
| | - Zahra-Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | |
Collapse
|
49
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [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: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
50
|
Wang L, Wang J, Xu A, Wei L, Pei M, Shen T, Xian X, Yang K, Fei L, Pan Y, Yang H, Wang X. Future embracing: exosomes driving a revolutionary approach to the diagnosis and treatment of idiopathic membranous nephropathy. J Nanobiotechnology 2024; 22:472. [PMID: 39118155 PMCID: PMC11312222 DOI: 10.1186/s12951-024-02633-y] [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: 03/09/2024] [Accepted: 06/13/2024] [Indexed: 08/10/2024] Open
Abstract
Membranous nephropathy (MN) is a leading cause of nephrotic syndrome in adults and is associated with high rates of end-stage renal disease. Early detection and precise interventions are crucial for improving patient prognosis and quality of life. However, the current diagnosis primarily relies on renal biopsies and traditional biomarkers, which have limitations. Additionally, targeted therapeutic strategies are lacking. Exosomes, small vesicles that facilitate intercellular communication, have emerged as potential noninvasive diagnostic markers due to their stability, diverse cargo, and rapid detectability. They also hold promise as carriers for gene and drug delivery, presenting innovative opportunities in renal disease prognosis and treatment. However, research on exosomes in the context of idiopathic membranous nephropathy (IMN) remains limited, with a focus on exploring urinary exosomes as IMN markers. In this review, we summarize the current status of MN diagnosis and treatment, highlight the fundamental characteristics of exosomes, and discuss recent advancements in their application to IMN diagnosis and therapy. We provide insights into the clinical prospects of exosomes in IMN and acknowledge potential challenges. This article aims to offer forward-looking insights into the future of exosome-mediated IMN diagnosis and treatment, indicating a revolutionary transformation in this field.
Collapse
Affiliation(s)
- Lin Wang
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jinxiang Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Guangdong, 518107, China
| | - Ao Xu
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lijuan Wei
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
| | - Ming Pei
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
| | - Tuwei Shen
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xian Xian
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kang Yang
- Nephrology Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Henan, 450099, China
| | - Lingyan Fei
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Yihang Pan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Guangdong, 518107, China.
| | - Hongtao Yang
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China.
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|