1
|
Wang B, Chen P, Li W, Chen Z. Exosomes in stroke management: A promising paradigm shift in stroke therapy. Neural Regen Res 2026; 21:6-22. [PMID: 39665811 PMCID: PMC12094539 DOI: 10.4103/nrr.nrr-d-24-00665] [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: 07/23/2024] [Revised: 07/27/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Effective treatment methods for stroke, a common cerebrovascular disease with a high mortality rate, are still being sought. Exosome therapy, a form of acellular therapy, has demonstrated promising efficacy in various diseases in animal models; however, there is currently insufficient evidence to guide the clinical application of exosome in patients with stroke. This article reviews the progress of exosome applications in stroke treatment. It aims to elucidate the significant potential value of exosomes in stroke therapy and provide a reference for their clinical translation. At present, many studies on exosome-based therapies for stroke are actively underway. Regarding preclinical research, exosomes, as bioactive substances with diverse sources, currently favor stem cells as their origin. Due to their high plasticity, exosomes can be effectively modified through various physical, chemical, and genetic engineering methods to enhance their efficacy. In animal models of stroke, exosome therapy can reduce neuroinflammatory responses, alleviate oxidative stress damage, and inhibit programmed cell death. Additionally, exosomes can promote angiogenesis, repair and regenerate damaged white matter fiber bundles, and facilitate the migration and differentiation of neural stem cells, aiding the repair process. We also summarize new directions for the application of exosomes, specifically the exosome intervention through the ventricular-meningeal lymphatic system. The review findings suggest that the treatment paradigm for stroke is poised for transformation.
Collapse
Affiliation(s)
- Bo Wang
- Department of Neurosurgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Pinzhen Chen
- Department of Radiology, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Wenyan Li
- Department of Neurosurgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| | - Zhi Chen
- Department of Neurosurgery, The First Hospital Affiliated to Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Mei Z, Li H, Huang C, Ma S, Li Y, Deng P, Zhou S, Qian A, Yang B, Li J. Extracellular vesicles from adipose-derived stromal/stem cells reprogram dendritic cells to alleviate rat TMJOA by transferring mitochondria. J Nanobiotechnology 2025; 23:389. [PMID: 40426246 PMCID: PMC12117725 DOI: 10.1186/s12951-025-03478-9] [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: 02/20/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) urgently needs regenerative therapies due to the limited effects of traditional treatments. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are considered a potent alternative for MSC therapy for the treatment of TMJOA. However, the specific mechanisms remain inadequately investigated. In this study, we explored how EVs from adipose-derived stromal/stem cells (ASCs) influence the TMJOA model triggered by Complete Freund's Adjuvant in rats and their impact on the state of dendritic cells (DCs) under pathological conditions. Subsequently, we conducted transcriptomic and metabolomic analyses to elucidate the specific mechanisms by which EVs affect DCs. Mechanistically, we demonstrate that EVs transferred functional mitochondria to DCs, which reverses their metabolic states. The internalized functional mitochondria from EVs activate the MAPK/ERK1/2/FoxO1/autophagy pathway, which causes the metabolic reprogramming of DCs and facilitates the achievement of therapeutic effects. These findings provide a mechanistic rationale for utilizing ASCs-EVs as cell-free alternatives to MSC transplantation in TMJOA therapy.
Collapse
Affiliation(s)
- Ziyi Mei
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Hanyue Li
- Department of Stomatology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chuling Huang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Shiyong Ma
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, The Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yuejia Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Pingmeng Deng
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Sha Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Aizhuo Qian
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Bin Yang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China.
- Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Changaei M, Azimzadeh Tabrizi Z, Karimi M, Kashfi SA, Koochaki Chahardeh T, Hashemi SM, Soudi S. From powerhouse to modulator: regulating immune system responses through intracellular mitochondrial transfer. Cell Commun Signal 2025; 23:232. [PMID: 40394666 PMCID: PMC12090700 DOI: 10.1186/s12964-025-02237-5] [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: 03/11/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Mitochondria are traditionally known as the cells' powerhouses; however, their roles go far beyond energy suppliers. They are involved in intracellular signaling and thus play a crucial role in shaping cells' destiny and functionality, including immune cells. Mitochondria can be actively exchanged between immune and non-immune cells via mechanisms such as nanotubes and extracellular vesicles. The mitochondria transfer from immune cells to different cells is associated with physiological and pathological processes, including inflammatory disorders, cardiovascular diseases, diabetes, and cancer. On the other hand, mitochondrial transfer from mesenchymal stem cells, bone marrow-derived stem cells, and adipocytes to immune cells significantly affects their functions. Mitochondrial transfer can prevent exhaustion/senescence in immune cells through intracellular signaling pathways and metabolic reprogramming. Thus, it is emerging as a promising therapeutic strategy for immune system diseases, especially those involving inflammation and autoimmune components. Transferring healthy mitochondria into damaged or dysfunctional cells can restore mitochondrial function, which is crucial for cellular energy production, immune regulation, and inflammation control. Also, mitochondrial transfer may enhance the potential of current therapeutic immune cell-based therapies such as CAR-T cell therapy.
Collapse
Affiliation(s)
- Mostafa Changaei
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Azimzadeh Tabrizi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mozhdeh Karimi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Adnan Kashfi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Tina Koochaki Chahardeh
- Department of Basic Sciences, Biology and Health, Faculty of Interdisciplinary Sciences and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Gao F, Pan L, Liu W, Chen J, Wang Y, Li Y, Liu Y, Hua Y, Li R, Zhang T, Zhu T, Jin F, Gao Y. Idiopathic pulmonary fibrosis microenvironment: Novel mechanisms and research directions. Int Immunopharmacol 2025; 155:114653. [PMID: 40222273 DOI: 10.1016/j.intimp.2025.114653] [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: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressive interstitial lung disease marked by increasing dyspnea and respiratory failure. The underlying mechanisms remain poorly understood, given the complexity of its pathogenesis. This review investigates the microenvironment of IPF to identify novel mechanisms and therapeutic avenues. Studies have revealed that various cell types, including alveolar epithelial cells, fibroblasts, myofibroblasts, and immune cells, are integral to disease progression, engaging in cellular stress responses and inflammatory regulation via signaling pathways such as TGF-β, Wnt, mTOR, and ROS. Non-coding RNAs, particularly miRNAs, are critical in IPF and may serve as diagnostic and prognostic biomarkers. Regarding treatment, mesenchymal stem cells (MSCs) and their extracellular vesicles (EVs) or non-vesicular derivatives offer promise by modulating immune responses, enhancing tissue repair, and inhibiting fibrosis. Additionally, alterations in the lung microbiota are increasingly recognized as a contributing factor to IPF progression, offering fresh insights into potential treatments. Despite the encouraging results of MSC-based therapies, the precise mechanisms and clinical applications remain subjects of ongoing research. This review emphasizes the significance of the IPF microenvironment and highlights the need for further exploration to develop effective therapies that could enhance patient outcomes.
Collapse
Affiliation(s)
- Fuguo Gao
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Wei Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jian Chen
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yifeng Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yan Li
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China; Department of Pulmonary and Critical Care Medicine, Shaanxi provincal people's hospital, Xi'an, 710068, China
| | - Yurou Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yiying Hua
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ruiqi Li
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Tongtong Zhang
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Ting Zhu
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Yongheng Gao
- Department of Pulmonary and Critical Care Medicine, Tangdu hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
5
|
Somova V, Jaborova N, Porubska B, Vasek D, Fikarova N, Prevorovsky M, Nahacka Z, Neuzil J, Krulova M. Mesenchymal stem cell-mediated mitochondrial transfer regulates the fate of B lymphocytes. Eur J Clin Invest 2025:e70073. [PMID: 40371939 DOI: 10.1111/eci.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Mitochondrial transfer is becoming recognized as an important immunomodulatory mechanism used by mesenchymal stem cells (MSCs) to influence immune cells. While effects on T cells and macrophages have been documented, the influence on B cells remains unexplored. This study investigates the modulation of B lymphocyte fate by MSC-mediated mitochondrial transfer. METHODS MSCs labelled with MitoTracker dyes or derived from mito::mKate2 transgenic mice were co-cultured with splenocytes. Flow cytometry assessed mitochondrial transfer, reactive oxygen species (ROS) levels, apoptosis and mitophagy. Glucose uptake was measured using the 2-NBDG assay. RNA sequencing analysed gene expression changes in CD19+ mitochondria recipients and nonrecipients. Pathway analysis identified affected processes. In an LPS-induced inflammation model, mito::mKate2 MSCs were administered, and B cells from different organs were analysed for mitochondrial uptake and phenotypic changes. MSC-derived mitochondria were also isolated to confirm uptake by FACS-sorted CD19+ cells. RESULTS MSCs transferred mitochondria to CD19+ cells, though less than to other immune cells. Transfer correlated with ROS levels and mitophagy induction. Mitochondria were preferentially acquired by activated B cells, as indicated by increased CD69 expression and glycolytic activity. Bidirectional transfer occurred, with immune cells exchanging dysfunctional mitochondria for functional ones. CD19+ recipients exhibited increased viability, proliferation and altered gene expression, with upregulated cell division genes and downregulated antigen presentation genes. In vivo, mitochondrial acquisition reduced B cell activation and inflammatory cytokine production. Pre-sorted B cells also acquired isolated mitochondria, exhibiting a similar anti-inflammatory phenotype. CONCLUSIONS These findings highlight mitochondrial trafficking as a key MSC-immune cell interaction mechanism with immunomodulatory therapeutic potential.
Collapse
Affiliation(s)
- Veronika Somova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Natalie Jaborova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Bianka Porubska
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Daniel Vasek
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Natalie Fikarova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Martin Prevorovsky
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Zuzana Nahacka
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jiri Neuzil
- Laboratory of Molecular Therapy, Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Magdalena Krulova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
6
|
Khan I, Ramzan F, Tayyab H, Damji KF. Rekindling Vision: Innovative Strategies for Treating Retinal Degeneration. Int J Mol Sci 2025; 26:4078. [PMID: 40362317 PMCID: PMC12072091 DOI: 10.3390/ijms26094078] [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: 03/07/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Retinal degeneration, characterized by the progressive loss of photoreceptors, retinal pigment epithelium cells, and/or ganglion cells, is a leading cause of vision impairment. These diseases are generally classified as inherited (e.g., retinitis pigmentosa, Stargardt disease) or acquired (e.g., age-related macular degeneration, diabetic retinopathy, glaucoma) ocular disorders that can lead to blindness. Available treatment options focus on managing symptoms or slowing disease progression and do not address the underlying causes of these diseases. However, recent advancements in regenerative medicine offer alternative solutions for repairing or protecting degenerated retinal tissue. Stem and progenitor cell therapies have shown great potential to differentiate into various retinal cell types and can be combined with gene editing, extracellular vesicles and exosomes, and bioactive molecules to modulate degenerative cellular pathways. Additionally, gene therapy and neuroprotective molecules play a crucial role in enhancing the efficacy of regenerative approaches. These innovative strategies hold the potential to halt the progression of retinal degenerative disorders, repair or replace damaged cells, and improve visual function, ultimately leading to a better quality of life for those affected.
Collapse
Affiliation(s)
- Irfan Khan
- Department of Ophthalmology and Visual Sciences, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Sindh, Pakistan;
- Centre for Regenerative Medicine and Stem Cells Research, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Sindh, Pakistan
- Department of Biological and Biomedical Sciences, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Sindh, Pakistan
| | - Faiza Ramzan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan;
| | - Haroon Tayyab
- Department of Ophthalmology and Visual Sciences, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Sindh, Pakistan;
| | - Karim F. Damji
- Department of Ophthalmology and Visual Sciences, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Sindh, Pakistan;
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
7
|
Wang Y, Cao Y, Xie W, Guo Y, Cai J, Huang T, Li P. Advances in clinical translation of stem cell-based therapy in neurological diseases. J Cereb Blood Flow Metab 2025; 45:600-616. [PMID: 39883811 PMCID: PMC11783424 DOI: 10.1177/0271678x251317374] [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: 10/19/2024] [Revised: 01/03/2025] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Stem cell-based therapies have raised considerable interest to develop regenerative treatment for neurological disorders with high disability. In this review, we focus on recent preclinical and clinical evidence of stem cell therapy in the treatment of degenerative neurological diseases and discuss different cell types, delivery routes and biodistribution of stem cell therapy. In addition, recent advances of mechanistic insights of stem cell therapy, including functional replacement by exogenous cells, immunomodulation and paracrine effects of stem cell therapies are also demonstrated. Finally, we also highlight the adjunction approaches that has been implemented to augment their reparative function, survival and migration to target specific tissue, including stem cell preconditioning, genetical engineering, co-transplantation and combined therapy.
Collapse
Affiliation(s)
- Yu Wang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yirong Cao
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Wanqing Xie
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Yunlu Guo
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Jiayi Cai
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Huang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Peiying Li
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| |
Collapse
|
8
|
Shi Y, Qin J, Yin E, Xu J, Chen Y, Tie K, Chen L. Mitochondria Isolated From Bone Mesenchymal Stem Cells Restrain Muscle Disuse Atrophy and Fatty Infiltration After Rotator Cuff Tears. Am J Sports Med 2025; 53:1171-1183. [PMID: 40088076 DOI: 10.1177/03635465251323001] [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: 03/17/2025]
Abstract
BACKGROUND Rotator cuff tears (RCTs) commonly lead to muscle atrophy, fibrosis, and fatty infiltration, complicating treatment. PURPOSE To investigate the use of mitochondria isolated from bone mesenchymal stem cells (BMSC-Mito) for mitigating complications after RCT, focusing on muscle protection. STUDY DESIGN Controlled laboratory study. METHODS RCTs were induced by transecting the tendons of the supraspinatus and infraspinatus in Sprague-Dawley rats. In vivo, 90 rats were randomized into 3 groups: sham (no intervention), RCTs treated with BMSC-Mito, and RCTs treated with phosphate-buffered saline. After 6 weeks of intramuscular injections of BMSC-Mito or phosphate-buffered saline, supraspinatus muscles were harvested for analysis. Evaluations included wet muscle weight, muscle fiber cross-sectional area, fibrosis, fatty infiltration, slow-fast myofiber types and muscle biomechanics, capillary density, mitochondria respiratory chain complex activity, adenosine triphosphate (ATP) concentration, oxidative stress, and mitochondrial ultrastructure. In vitro experiments utilized primary rat skeletal muscle cells pretreated with rhodamine 6G to induce mitochondrial dysfunction, assessing the effects of BMSC-Mito on cell viability, mitochondrial membrane potential, and oxidative stress levels. RESULTS BMSC-Mito can be effectively transplanted into muscles and integrated into the local mitochondrial network. After RCT, the supraspinatus showed significant mass loss, reduced fiber cross-sectional area, fatty infiltration, and a shift from slow to fast myofiber types, which negatively affected muscle biomechanics. These changes were reversed by BMSC-Mito. BMSC-Mito also preserved vascularity (CD31 and α-SMA) impaired by RCT. Additionally, BMSC-Mito notably improved disuse-induced mitochondrial changes, leading to increased mitochondrial number and COX IV expression; furthermore, BMSC-Mito protected mitochondria morphology and enhanced cytosolic superoxide dismutase activity. This treatment also improved mitochondria respiratory chain complex activity and ATP concentration, reducing oxidative stress. In vitro, BMSC-Mito treatment effectively maintained the mitochondrial membrane potential of skeletal muscle cells, improved cell viability, and restored its mitochondrial function and ATP levels. CONCLUSION These findings suggest that BMSC-Mito might play a role in preventing muscle atrophy and fatty infiltration after RCT through the protection of mitochondrial function and the promotion of angiogenesis. CLINICAL RELEVANCE BMSC-Mito present a promising therapeutic approach for addressing rotator cuff muscle degeneration.
Collapse
Affiliation(s)
- Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Qin
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Enzhi Yin
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Kai Tie
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Xiong Z, Chen P, Wang Z, Yao L, Yuan M, Liu P, Sun M, Shu K, Jiang Y. Human umbilical cord-derived mesenchymal stem cells attenuate liver fibrosis by inhibiting hepatocyte ferroptosis through mitochondrial transfer. Free Radic Biol Med 2025; 231:163-177. [PMID: 40023296 DOI: 10.1016/j.freeradbiomed.2025.02.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Liver fibrosis is a reversible dynamic pathological process induced by chronic liver injury. Without intervention, liver fibrosis can progress to become cirrhosis, liver failure, or hepatocellular carcinoma, thus posing a high global health burden. Therefore, effective therapies for liver fibrosis are urgently required. Although transplantation of mesenchymal stem cells (MSCs) has significant value as a treatment strategy for liver damage, the underlying mechanisms remain unclear. Chronic liver injury progression is significantly influenced by hepatocyte ferroptosis, and targeting ferroptosis is emerging as a potential treatment strategy for liver fibrosis. Here, we showed that the infusion of human umbilical cord-derived MSCs (hUC-MSCs) alleviated TAA-induced liver fibrosis, improved liver functionality, and decreased ferroptosis in mice. hUC-MSCs inhibit ferroptosis-related mitochondrial damage and lipid peroxidation in AML12 cells in vitro. Mechanistically, under oxidative stress, hUC-MSCs transfer healthy mitochondria to damaged hepatocytes through tunneling nanotubes (TNTs). Cytochalasin D (CytoD), an inhibitor of TNT formation, abrogated the protective effects of hUC-MSCs against ferroptosis. This research emphasizes the ability of hUC-MSCs to serve as a promising treatment for liver fibrosis via mitochondrial transfer through TNTs.
Collapse
Affiliation(s)
- Zhiyu Xiong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Ping Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Zheng Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Mengqin Yuan
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, China.
| | - Pingji Liu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Muhua Sun
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Kan Shu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
10
|
Gao X, Chen Y, Wang J, Xu J, Wan H, Li X, Shi Y. Mitochondria-Rich Extracellular Vesicles From Bone Marrow Stem Cells Mitigate Muscle Degeneration in Rotator Cuff Tears in a Rat Model Through Macrophage M2 Phenotype Conversion. Arthroscopy 2025:S0749-8063(25)00229-4. [PMID: 40147598 DOI: 10.1016/j.arthro.2025.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE To investigate the protective effects of extracellular vesicles derived from bone marrow stem cells (BMSC-EVs) on muscle degeneration in a rat model of rotator cuff tendon and suprascapular nerve (SSN) transection (termed the RCT-SSN model), focusing on mitochondrial transfer. METHODS The EVs were identified and characterized. The RCT-SSN model was established by transecting the supraspinatus, infraspinatus tendons, and suprascapular nerve. Ninety-six rats were divided into 4 groups (n = 24 each): sham surgery, RCT-SSN treated with BMSC-EVs, RCT-SSN treated with EVs from rhodamine 6G-pretreated BMSCs (Rho-EVs), or phosphate-buffered saline. Intramuscular injections were administered every 2 weeks. After 12 weeks, supraspinatus muscles were analyzed for atrophy, fibrosis, oxidative stress, macrophage phenotypes, serum cytokines, and mitochondrial characteristics. In vitro experiments included EVs tracking in macrophages, macrophage phenotype characterization, and inflammatory cytokine profiling. RESULTS BMSC-EVs and Rho-EVs displayed similar morphology, but only BMSC-EVs contained functional mitochondria. BMSC-EVs significantly reduced muscle weight loss (0.047 ± 0.010% vs 0.145 ± 0.013%, P < .001), increased muscle fiber cross-sectional area (2037 ± 231.9 μm2 vs 527.9 ± 92.01 μm2, P < .001), and decreased fibrosis (12.09 ± 3.31% vs 25.69 ± 4.84%, P < .001) compared with phosphate-buffered saline. BMSC-EVs enhanced superoxide dismutase activity (93.3 ± 11.8 U/mg protein vs 53.4 ± 8.0 U/mg protein, P < .001), improved mitochondrial function, density and structure, and induced an anti-inflammatory macrophage shift, suppressing proinflammatory cytokines in vitro and in vivo. Rho-EVs showed no such effects. CONCLUSIONS This study showed that transecting the supraspinatus, infraspinatus tendons, and suprascapular nerve in a rat model induced muscle degeneration and fibrosis. BMSC-EVs, but not Rho-EVs, mitigated these effects by promoting an anti-inflammatory macrophage phenotype and protecting mitochondrial function through mitochondrial transfer. CLINICAL RELEVANCE Mitochondrial transfer via BMSC-EVs may offer a therapeutic strategy to prevent muscle degeneration in patients with rotator cuff tear.
Collapse
Affiliation(s)
- Xing Gao
- Animal Experimental Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, China
| | - Jingzeng Wang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hu Wan
- Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao Li
- Priority Medical Department, General Hospital of Central Theater command, Wuhan, China
| | - Yulong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
11
|
Li X, Su J, Liu X, Lu W, Deng Z. Mitochondria derived from Stem cells modulated the biological behavior of monocyte-macrophages and inhibited inflammatory bone resorption. BMC Musculoskelet Disord 2025; 26:286. [PMID: 40121414 PMCID: PMC11929288 DOI: 10.1186/s12891-025-08529-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND The transfer of mitochondria from stem cells effectively attenuates the viability of inflammatory cells. However, there is a paucity of research supporting the inhibitory effect of stem cells on inflammatory bone resorption through mitochondrial transfer. METHODS Mouse bone resorption models were established to investigate the impact of stem cell-derived mitochondria. Stem cells, stem cell-derived mitochondria and exosomes were injected into the animal models for experimental research. Healthy mice and mice with bone resorption were included as the control groups. The mitochondrial transfer and bone resorption of mice calvaria were evaluated by immunofluorescence, gross morphology, micro-computed tomography (micro-CT), immunohistochemical staining. Monocyte-macrophages were incubated with stem cell-derived mitochondria as experimental group. Monocyte-macrophages and activated monocyte-macrophages cultured separately served as the control groups. The mitochondrial transfer and biological behavior of monocyte-macrophages were evaluated by immunofluorescence, enzyme-linked immunosorbent assay (ELISA), Multiskan FC, and histochemical staining. RESULTS Stem cell-derived mitochondria were successfully transferred to monocyte-macrophages. In vivo, local injection of stem cells, mitochondria, and exosomes effectively mitigated inflammatory cell infiltration, suppressed osteoclast maturation, and demonstrated a higher relative bone volume in mouse bone resorption models compared to the negative control group. In vitro, the co-incubation of mitochondria effectively suppressed the secretion of inflammatory cytokines, proliferation, fusion, and osteoclastogenesis in monocyte-macrophages compared to the control groups. CONCLUSIONS The modulation of monocyte-macrophages biological behaviors by stem cells may occur through the transfer of mitochondria, thereby mitigating inflammatory bone resorption.
Collapse
Affiliation(s)
- Xingfu Li
- Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, 518035, Guangdong, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Jingyue Su
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325005, Zhejiang, China
- Geriatrics Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325005, Zhejiang, China
| | - Xiang Liu
- Department of Orthopedics, Heilongjiang Provincial Hospital, Harbin, 150036, Heilongjiang, China
| | - Wei Lu
- Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, 518035, Guangdong, China.
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China.
| | - Zhenhan Deng
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325005, Zhejiang, China.
- Geriatrics Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325005, Zhejiang, China.
| |
Collapse
|
12
|
Yan J, Yang T, Ma S, Li D, Hu C, Tan J. Macrophage-derived mitochondria-rich extracellular vesicles aggravate bone loss in periodontitis by disrupting the mitochondrial dynamics of BMSCs. J Nanobiotechnology 2025; 23:208. [PMID: 40075447 PMCID: PMC11905510 DOI: 10.1186/s12951-025-03178-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Periodontitis is the leading cause of tooth loss in adults due to progressive bone destruction, which is closely related to the dysfunction of bone mesenchymal stem cells (BMSCs). Existing evidence suggests that mitochondrial disorders are associated with periodontitis. However, whether mitochondrial dysregulation contributes to the osteogenic impairment of BMSCs and the underlying mechanisms remain unclear. Macrophages have been shown to communicate extensively with BMSCs in periodontitis. Recent studies have reported a novel manner of cellular communication in which mitochondria-rich extracellular vesicles(MEVs) transfer mitochondria from parent cells to recipient cells, playing a role in both physiological and pathological conditions. Therefore, we aimed to investigate the role of MEVs in orchestrating the crosstalk between macrophages and BMSCs in periodontitis to formulate management strategies for bone loss. RESULTS Our results revealed that macrophages underwent significant mitochondrial dysfunction and inflammation in periodontitis and that MEVs derived from these macrophages played a role in alveolar bone destruction. Furthermore, cell imaging showed that inflammatory macrophages packaged numerous damaged mitochondria into MEVs, and the entry of these impaired mitochondria into BMSCs disrupted mitochondrial dynamics and hindered donut-shaped mitochondria formation, leading to osteogenic dysfunction. Proteomic analysis revealed that the proteins enriched in macrophage-derived MEVs were largely related to mitochondria and the formation and transport of vesicles. Additionally, we found that MEVs from macrophages significantly increased lipocalin 2 (LCN2) in BMSCs in periodontitis and that LCN2 perturbed mitochondrial morphological changes in BMSCs by inducing the degradation of OMA1 and accumulation of OPA1, resulting in osteogenesis impairment in BMSCs. Inhibition of LCN2 rescued the osteogenic dysfunction of BMSCs and alveolar bone loss in periodontitis. CONCLUSIONS The transfer of mitochondria to BMSCs via MEVs exacerbates alveolar bone resorption through LCN2/OMA1/OPA1 signaling in periodontitis. Inhibition of LCN2 alleviates inflammatory bone loss, suggesting a promising therapeutic strategy for periodontitis.
Collapse
Affiliation(s)
- Jiayin Yan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tian Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
| | - Siyuan Ma
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
| | - Danfeng Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
| | - Cheng Hu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiali Tan
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China.
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
13
|
Chang MM, Chu DT, Lin SC, Lee JS, Vu TD, Vu HT, Ramasamy TS, Lin SP, Wu CC. Enhanced mitochondrial function and delivery from adipose-derived stem cell spheres via the EZH2-H3K27me3-PPARγ pathway for advanced therapy. Stem Cell Res Ther 2025; 16:129. [PMID: 40069892 PMCID: PMC11899936 DOI: 10.1186/s13287-025-04164-1] [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: 09/25/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Microenvironmental alterations induce significant genetic and epigenetic changes in stem cells. Mitochondria, essential for regenerative capabilities, provide the necessary energy for stem cell function. However, the specific roles of histone modifications and mitochondrial dynamics in human adipose-derived stem cells (ASCs) during morphological transformations remain poorly understood. In this study, we aim to elucidate the mechanisms by which ASC sphere formation enhances mitochondrial function, delivery, and rescue efficiency. METHODS ASCs were cultured on chitosan nano-deposited surfaces to form 3D spheres. Mitochondrial activity and ATP production were assessed using MitoTracker staining, Seahorse XF analysis, and ATP luminescence assays. Single-cell RNA sequencing, followed by Ingenuity Pathway Analysis (IPA), was conducted to uncover key regulatory pathways, which were validated through molecular techniques. Pathway involvement was confirmed using epigenetic inhibitors or PPARγ-modulating drugs. Mitochondrial structural integrity and delivery efficiency were evaluated after isolation. RESULTS Chitosan-induced ASC spheres exhibited unique compact mitochondrial morphology, characterized by condensed cristae, enhanced mitochondrial activity, and increased ATP production through oxidative phosphorylation. High expressions of mitochondrial complex I genes and elevated levels of mitochondrial complex proteins were observed without an increase in reactive oxygen species (ROS). Epigenetic modification of H3K27me3 and PPARγ involvement were discovered and confirmed by inhibiting H3K27me3 with the specific EZH2 inhibitor GSK126 and by adding the PPARγ agonist Rosiglitazone (RSG). Isolated mitochondria from ASC spheres showed improved structural stability and delivery efficiency, suppressed the of inflammatory cytokines in LPS- and TNFα-induced inflamed cells, and rescued cells from damage, thereby enhancing function and promoting recovery. CONCLUSION Enhancing mitochondrial ATP production via the EZH2-H3K27me3-PPARγ pathway offers an alternative strategy to conventional cell-based therapies. High-functional mitochondria and delivery efficiency show significant potential for regenerative medicine applications.
Collapse
Affiliation(s)
- Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Dinh Toi Chu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Sheng-Che Lin
- Division of Plastic and Reconstructive Surgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, 70965, Taiwan
| | - Jung-Shun Lee
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, 701401, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Thuy Duong Vu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Hue Thi Vu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Shau-Ping Lin
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, 10672, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan.
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101, Taiwan.
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
14
|
Wang R, Fu J, He J, Wang X, Xing W, Liu X, Yao J, Ye Q, He Y. Apoptotic mesenchymal stem cells and their secreted apoptotic extracellular vesicles: therapeutic applications and mechanisms. Stem Cell Res Ther 2025; 16:78. [PMID: 39985021 PMCID: PMC11846181 DOI: 10.1186/s13287-025-04211-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/30/2025] [Indexed: 02/23/2025] Open
Abstract
Mesenchymal stem cells (MSCs), an accessible and less ethically controversial class of adult stem cells, have demonstrated significant efficacy in treating a wide range of diseases in both the preclinical and clinical phases. However, we do not yet have a clear understanding of the mechanisms by which MSCs exert their therapeutic effects in vivo. We found that the transplanted MSCs go an apoptotic fate within 24 h in vivo irrespective of the route of administration. Still, the short-term survival of MSCs do not affect their long-term therapeutic efficacy. An increasing number of studies have demonstrated that transplantation of apoptotic MSCs (ApoMSCs) show similar or even better efficacy than viable MSCs, including a variety of preclinical disease models such as inflammatory diseases, skin damage, bone damage, organ damage, etc. Although the exact mechanism has yet to be explored, recent studies have shown that transplanted MSCs undergo apoptosis in vivo and are phagocytosed by phagocytes, thereby exerting immunomodulatory effects. The apoptotic extracellular vesicles secreted by ApoMSCs (MSC-ApoEVs) play a significant role in promoting immunomodulation, endogenous stem cell regeneration, and angiogenesis due to their apoptotic properties and inheritance of molecular characteristics from their parental MSCs. On this basis, this review aims to deeply explore the therapeutic applications and mechanisms of ApoMSCs and their secretion of MSC-ApoEVs, as well as the challenges they face.
Collapse
Affiliation(s)
- Ruoxuan Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jiao Fu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jihui He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xinxin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Wenbo Xing
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaojing Liu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Juming Yao
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China.
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China.
- Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Zhao J, Fang W, Gao Y, Chen J, Wang G, Gu J. iPSC-derived exosomes as amphotericin B carriers: a promising approach to combat cryptococcal meningitis. Front Microbiol 2025; 16:1531425. [PMID: 39996080 PMCID: PMC11847882 DOI: 10.3389/fmicb.2025.1531425] [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: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Background Cryptococcal meningitis (CM) is a significant global health issue, particularly affecting individuals with HIV. Amphotericin B (AmB) serves as the cornerstone treatment for CM; however, its clinical application is restricted due to limited penetration of the blood-brain barrier and associated nephrotoxicity. Objective This study investigates the use of exosomes derived from induced pluripotent stem cells (iPSC-Exos) as carriers for AmB in treating CM, aiming to enhance therapeutic efficacy and safety and reduce AmB toxicity. Methods Exosomes were extracted from iPSC culture supernatants using ultrafiltration and ultracentrifugation. Their morphology and size were analyzed using transmission electron microscopy (TEM) and nanoparticle flow cytometry (nFCM). Purity was confirmed by Western blotting for markers CD9, CD63, and TSG101. AmB was loaded into iPSC-Exos using a co-incubation method. The cytotoxicity of the iPSC-Exo/AmB complex was evaluated on HEK 293 T and RAW264.7 cells using the CCK-8 assay, while apoptosis was assessed using live/dead cell staining and flow cytometry. The hemolytic effects were tested using rabbit red blood cells. In a C57BL/6 J mouse model of cryptococcal infection, treatment groups (AmB, iPSC-Exo/AmB, and iPSC-Exo) were administered corresponding drugs, with blood and brain samples collected for analysis. The minimum inhibitory concentration (MIC) of iPSC-Exo/AmB and conventional AmB against Cryptococcus was determined. Results The iPSC-Exo/AmB complex exhibited reduced cytotoxicity in vitro and decreased AmB-induced renal and hepatic toxicity in vivo. Its MIC against Cryptococcus was over eight times lower than conventional AmB, significantly reducing fungal burden in the mouse brain and lowering serum inflammatory factors. Conclusion The iPSC-Exo/AmB complex is a promising therapeutic strategy that enhances AmB efficacy while reducing toxicity, offering new hope for treating CM and other refractory fungal infections of the central nervous system.
Collapse
Affiliation(s)
- Jingyu Zhao
- Department of Dermatology, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Fang
- Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yangjie Gao
- Department of Dermatology, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiquan Chen
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People’s Hospital, School of Medicine Tongji University, Shanghai, China
| | - Julin Gu
- Department of Dermatology, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Li X, Lu W, Ni L, Su J, Wang D, Deng Z. Mitochondria-rich extracellular vesicles derived from the culture supernatant of human synovial Fluid-derived mesenchymal stem cells Inhibited senescence of Stressed/inflammatory Licensed chondrocytes and Delayed Osteoarthritis progression. Int Immunopharmacol 2025; 147:113954. [PMID: 39756162 DOI: 10.1016/j.intimp.2024.113954] [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: 08/23/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Mitochondrial dysfunction induces chondrocyte senescence, thereby precipitating articular cartilage (AC) degeneration in the pathogenesis of osteoarthritis (OA). Although the transfer of mitochondria from mesenchymal stem cells (MSCs) to host cells and their potential protective role have been demonstrated, whether MSCs can alleviate chondrocyte mitochondrial dysfunction or reverse OA progression remains unclear. METHODS A mitochondrial tracer was used to investigate the transfer of mitochondria-rich extracellular vesicles (MEV) derived from the culture supernatant of human synovial fluid-derived mesenchymal stem cells (hSF-MSCs). Human articular chondrocytes (hACs) impaired by oxidative stress co-incubated with MEV were used for experimental research in vitro. Healthy hACs and stressed hACs were cultured separately acting as the control groups. The MEV was injected into the OA rats' knee joint serving as experimental group. Healthy and OA rats were served as the control groups. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blot (WB), enzyme- linked immunosorbent assay (ELISA), flow cytometry (FC), immunofluorescence (IF), fluorescence spectrophotometer (FS), immunohistochemistry (IHC) and other methods are used to analyze the effect of MEV on hACs and OA progression. RESULTS MEV derived from hSF-MSCs could transfer into hACs. Compared to the negative control group, co-incubation with MEV resulted in a significant down-regulation of oxidative stress markers and senescence-associated proteins in hACs, while improved mitochondrial function of hACs. Moreover, the MEV could traverse the dense interstitial layer and migrate towards the deeper cartilage, while intra-articular injection of MEV could effectively attenuate AC degeneration. CONCLUSION The transfer of MEV derived from hSF-MSCs represents a promising strategy for safeguarding AC, thereby offering a potential avenue and mechanism for the treatment of OA.
Collapse
Affiliation(s)
- Xingfu Li
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, Guangdong, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, Guangdong, China
| | - Wei Lu
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, Guangdong, China
| | - Linying Ni
- Department of Orthopedics, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Jingyue Su
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325005, Zhejiang, China; Geriatrics Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Daping Wang
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen 518035, Guangdong, China.
| | - Zhenhan Deng
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325005, Zhejiang, China; Geriatrics Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
17
|
Tang H, Wang H, Gan Z, Ding Z, Yu Q. Engineering the Hydrophilic-Hydrophobic Interface of Polymeric Micelles by Cationic Blocks for Enhanced Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:69011-69027. [PMID: 39639482 DOI: 10.1021/acsami.4c17024] [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: 12/07/2024]
Abstract
The cationic surface charge critically influences the biological functions and therapeutic outcomes of the cancer nanomedicines. However, the basic correlation between the cationic group categories and their therapeutic efficacy has not been elucidated. In this study, cationic polymeric nanoparticles with amino groups (primary, tertiary, and quaternary amines) as the single variable were leveraged to investigate the various effects of amino species for enhanced antitumor chemotherapy. The nanoparticles were constructed from a series of triblock polymers with varying cationic repeating units at the hydrophilic-hydrophobic interface. Our results suggested that quaternary ammonium outperforms its primary and tertiary counterparts in destroying mitochondrial membranes to induce apoptosis, penetrating deep inside the tumor tissue, and damaging tumor vasculatures. As a result, we were able to effectively inhibit tumor growth in mice by a quaternary ammonium conjugate without causing significant toxicity. Our work demonstrated that the chemical structures played vital roles in regulating their biological functions and provided valuable information for designing cationic drug delivery systems.
Collapse
Affiliation(s)
- Hao Tang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology; Shenzhen, Guangdong 518055, P. R. China
| | - Hanbing Wang
- The State Key Laboratory of Organic Inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Zhihua Gan
- The State Key Laboratory of Organic Inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Zhenshan Ding
- Department of Urology, China-Japan Friendship Hospital, Beijing 100029, P. R. China
| | - Qingsong Yu
- The State Key Laboratory of Organic Inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
18
|
Li H, Sun W, Gong W, Han Y. Transfer and fates of damaged mitochondria: role in health and disease. FEBS J 2024; 291:5342-5364. [PMID: 38545811 DOI: 10.1111/febs.17119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/27/2024] [Accepted: 03/04/2024] [Indexed: 12/19/2024]
Abstract
Intercellular communication is pivotal in mediating the transfer of mitochondria from donor to recipient cells. This process orchestrates various biological functions, including tissue repair, cell proliferation, differentiation and cancer invasion. Typically, dysfunctional and depolarized mitochondria are eliminated through intracellular or extracellular pathways. Nevertheless, increasing evidence suggests that intercellular transfer of damaged mitochondria is associated with the pathogenesis of diverse diseases. This review investigates the prevalent triggers of mitochondrial damage and the underlying mechanisms of mitochondrial transfer, and elucidates the role of directional mitochondrial transfer in both physiological and pathological contexts. Additionally, we propose potential previously unknown mechanisms mediating mitochondrial transfer and explore their prospective roles in disease prevention and therapy.
Collapse
Affiliation(s)
- Hanbing Li
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Weiyun Sun
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenwen Gong
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yubing Han
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Belousova E, Salikhova D, Maksimov Y, Nebogatikov V, Sudina A, Goldshtein D, Ustyugov A. Proposed Mechanisms of Cell Therapy for Alzheimer's Disease. Int J Mol Sci 2024; 25:12378. [PMID: 39596443 PMCID: PMC11595163 DOI: 10.3390/ijms252212378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder characterized by mitochondria dysfunction, accumulation of beta-amyloid plaques, and hyperphosphorylated tau tangles in the brain leading to memory loss and cognitive deficits. There is currently no cure for this condition, but the potential of stem cells for the therapy of neurodegenerative pathologies is actively being researched. This review discusses preclinical and clinical studies that have used mouse models and human patients to investigate the use of novel types of stem cell treatment approaches. The findings provide valuable insights into the applications of stem cell-based therapies and include the use of neural, glial, mesenchymal, embryonic, and induced pluripotent stem cells. We cover current studies on stem cell replacement therapy where cells can functionally integrate into neural networks, replace damaged neurons, and strengthen impaired synaptic circuits in the brain. We address the paracrine action of stem cells acting via secreted factors to induce neuroregeneration and modify inflammatory responses. We focus on the neuroprotective functions of exosomes as well as their neurogenic and synaptogenic effects. We look into the shuttling of mitochondria through tunneling nanotubes that enables the transfer of healthy mitochondria by restoring the normal functioning of damaged cells, improving their metabolism, and reducing the level of apoptosis.
Collapse
Affiliation(s)
- Ekaterina Belousova
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
| | - Diana Salikhova
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
- Research Institute of Molecular and Cellular Medicine of the Medical Institute Peoples’ Friendship, University of Russia, Moscow 117198, Russia
| | - Yaroslav Maksimov
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
- Research Institute of Molecular and Cellular Medicine of the Medical Institute Peoples’ Friendship, University of Russia, Moscow 117198, Russia
| | - Vladimir Nebogatikov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry of the Russian Academy of Sciences, Chernogolovka 142432, Russia;
| | - Anastasiya Sudina
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
- Research Institute of Molecular and Cellular Medicine of the Medical Institute Peoples’ Friendship, University of Russia, Moscow 117198, Russia
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, Moscow 115522, Russia; (E.B.); (D.S.); (Y.M.); (A.S.); (D.G.)
| | - Aleksey Ustyugov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry of the Russian Academy of Sciences, Chernogolovka 142432, Russia;
| |
Collapse
|
20
|
Hazrati A, Mirarefin SMJ, Malekpour K, Rahimi A, Khosrojerdi A, Rasouli A, Akrami S, Soudi S. Mesenchymal stem cell application in pulmonary disease treatment with emphasis on their interaction with lung-resident immune cells. Front Immunol 2024; 15:1469696. [PMID: 39582867 PMCID: PMC11581898 DOI: 10.3389/fimmu.2024.1469696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 11/26/2024] Open
Abstract
Due to the vital importance of the lungs, lung-related diseases and their control are very important. Severe inflammatory responses mediated by immune cells were among the leading causes of lung tissue pathology and damage during the COVID-19 pandemic. In addition, uncontrolled immune cell responses can lead to lung tissue damage in other infectious and non-infectious diseases. It is essential to control immune responses in a way that leads to homeostasis. Immunosuppressive drugs only suppress inflammatory responses and do not affect the homeostasis of reactions. The therapeutic application of mesenchymal stem cells (MSCs), in addition to restoring immune homeostasis, can promote the regeneration of lung tissue through the production of growth factors and differentiation into lung-related cells. However, the communication between MSCs and immune cells after treatment of pulmonary diseases is essential, and investigating this can help develop a clinical perspective. Different studies in the clinical phase showed that MSCs can reverse fibrosis, increase regeneration, promote airway remodeling, and reduce damage to lung tissue. The proliferation and differentiation potential of MSCs is one of the mechanisms of their therapeutic effects. Furthermore, they can secrete exosomes that affect the function of lung cells and immune cells and change their function. Another important mechanism is that MSCs reduce harmful inflammatory responses through communication with innate and adaptive immune cells, which leads to a shift of the immune system toward regulatory and hemostatic responses.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arezou Rahimi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Arezou Khosrojerdi
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Ashkan Rasouli
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Susan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
21
|
Pal-Ghosh S, Karpinski BA, Datta-Majumdar H, Datta S, Dimri S, Hally J, Wehmeyer H, Stepp MA. Mechanisms Regulating Mitochondrial Transfer in Human Corneal Epithelial Cells. Invest Ophthalmol Vis Sci 2024; 65:10. [PMID: 39504055 PMCID: PMC11549930 DOI: 10.1167/iovs.65.13.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/23/2024] [Indexed: 11/11/2024] Open
Abstract
Purpose The intraepithelial corneal nerves (ICNs) innervating the cornea are essential to corneal epithelial cell homeostasis. Rho-associated kinase (ROCK) inhibitors (RIs) have been reported to play roles in neuron survival after injury and in mitochondrial transfer between corneal epithelial cells. In this study, the mechanisms human corneal limbal epithelial (HCLE) cells use to control intercellular mitochondrial transfer are assessed. Methods Mitotracker and AAV1 mitotag eGFPmCherry were used to allow us to study mitochondrial transfer between HCLE cells and neurons in co-cultures and in HCLE cultures. A mitochondrial transfer assay was developed using HCLE cells to quantify the impact of cell stress and inhibition of phagocytosis, gap junctions, and ROCK on mitochondrial transfer, cell adhesion, migration, matrix deposition, and mitochondrial content. Results Bidirectional mitochondrial transfer occurs between HCLE cells and neurons. Mitochondrial transfer among HCLE cells is inhibited when gap junction function is reduced and enhanced by acid stress and by inhibition of either phagocytosis or ROCK. Media conditioned by RI-treated cells stimulates cell adhesion and mitochondrial transfer. Conclusions Maximal mitochondrial transfer takes place when gap junctions are functional, when ROCK and phagocytosis are inhibited, and when cells are stressed by low pH media. Treatments that reduce mitochondrial content increase HCLE cell mitochondrial transfer. ROCK inhibition in co-cultures causes the release and adhesion of mitochondria to substrates where they can be engulfed by migrating HCLE cells and growing axons and their growth cones.
Collapse
Affiliation(s)
- Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Beverly A. Karpinski
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Himani Datta-Majumdar
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Soneha Datta
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Shelly Dimri
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Jordan Hally
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Hugo Wehmeyer
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
- Department of Ophthalmology, GW School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
22
|
Tian G, Yin H, Zheng J, Yu R, Ding Z, Yan Z, Tang Y, Wu J, Ning C, Yuan X, Liao C, Sui X, Zhao Z, Liu S, Guo W, Guo Q. Promotion of osteochondral repair through immune microenvironment regulation and activation of endogenous chondrogenesis via the release of apoptotic vesicles from donor MSCs. Bioact Mater 2024; 41:455-470. [PMID: 39188379 PMCID: PMC11347043 DOI: 10.1016/j.bioactmat.2024.07.034] [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: 04/24/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024] Open
Abstract
Utilizing transplanted human umbilical cord mesenchymal stem cells (HUMSCs) for cartilage defects yielded advanced tissue regeneration, but the underlying mechanism remain elucidated. Early after HUMSCs delivery to the defects, we observed substantial apoptosis. The released apoptotic vesicles (apoVs) of HUMSCs promoted cartilage regeneration by alleviating the chondro-immune microenvironment. ApoVs triggered M2 polarization in macrophages while simultaneously facilitating the chondrogenic differentiation of endogenous MSCs. Mechanistically, in macrophages, miR-100-5p delivered by apoVs activated the MAPK/ERK signaling pathway to promote M2 polarization. In MSCs, let-7i-5p delivered by apoVs promoted chondrogenic differentiation by targeting the eEF2K/p38 MAPK axis. Consequently, a cell-free cartilage regeneration strategy using apoVs combined with a decellularized cartilage extracellular matrix (DCM) scaffold effectively promoted the regeneration of osteochondral defects. Overall, new mechanisms of cartilage regeneration by transplanted MSCs were unconcealed in this study. Moreover, we provided a novel experimental basis for cell-free tissue engineering-based cartilage regeneration utilizing apoVs.
Collapse
Affiliation(s)
- Guangzhao Tian
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Han Yin
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinxuan Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Rongcheng Yu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Zhengang Ding
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zineng Yan
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Yiqi Tang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Jiang Wu
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Chao Ning
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Xun Yuan
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Chenxi Liao
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, China
| | - Xiang Sui
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zhe Zhao
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shuyun Liu
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Weimin Guo
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital Sun Yat-Sen University, Guangzhou, 510080, China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, China
- Institute of Orthopedies, Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, 51 Fucheng Road, Haidian District, Beijing, 100142, China
| |
Collapse
|
23
|
Luo H, Lai Y, Tang W, Wang G, Shen J, Liu H. Mitochondrial transplantation: a promising strategy for treating degenerative joint diseases. J Transl Med 2024; 22:941. [PMID: 39407249 PMCID: PMC11475785 DOI: 10.1186/s12967-024-05752-0] [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: 08/19/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
The prevalence of age-related degenerative joint diseases, particularly intervertebral disc degeneration and osteoarthritis, is increasing, thereby posing significant challenges for the elderly population. Mitochondrial dysfunction is a critical factor in the etiology and progression of these disorders. Therapeutic interventions that incorporate mitochondrial transplantation exhibit considerable promise by increasing mitochondrial numbers and improving their functionality. Existing evidence suggests that exogenous mitochondrial therapy improves clinical outcomes for patients with degenerative joint diseases. This review elucidates the mitochondrial abnormalities associated with degenerative joint diseases and examines the mechanisms of mitochondrial intercellular transfer and artificial mitochondrial transplantation. Furthermore, therapeutic strategies for mitochondrial transplantation in degenerative joint diseases are synthesized, and the concept of engineered mitochondrial transplantation is proposed.
Collapse
Affiliation(s)
- Hong Luo
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yue Lai
- Department of Orthopedics, Affiliated Hospital of Guangdong medical University, zhanjiang, 524000, China
| | - Weili Tang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Guoyou Wang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Jianlin Shen
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
24
|
Zhao M, Wang J, Zhu S, Wang M, Chen C, Wang L, Liu J. Mitochondrion-based organellar therapies for central nervous system diseases. Cell Commun Signal 2024; 22:487. [PMID: 39390521 PMCID: PMC11468137 DOI: 10.1186/s12964-024-01843-z] [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: 06/26/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
As most traditional drugs used to treat central nervous system (CNS) diseases have a single therapeutic target, many of them cannot treat complex diseases or diseases whose mechanism is unknown and cannot effectively reverse the root changes underlying CNS diseases. This raises the question of whether multiple functional components are involved in the complex pathological processes of CNS diseases. Organelles are the core functional units of cells, and the replacement of damaged organelles with healthy organelles allows the multitargeted and integrated modulation of cellular functions. The development of therapies that target independent functional units in the cell, specifically, organelle-based therapies, is rapidly progressing. This article comprehensively discusses the pathogenesis of mitochondrial homeostasis disorders, which involve mitochondria, one of the most important organelles in CNS diseases, and the machanisms of mitochondrion-based therapies, as well as current preclinical and clinical studies on the efficacy of therapies targeting mitochondrial to treat CNS diseases, to provide evidence for use of organelle-based treatment strategies in the future.
Collapse
Affiliation(s)
- Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Shuaiyu Zhu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Meina Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Chong Chen
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian City, Liaoning Province, 116011, P.R. China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian City, Liaoning Province, 116023, P.R. China.
| |
Collapse
|
25
|
Ore A, Angelastro JM, Giulivi C. Integrating Mitochondrial Biology into Innovative Cell Therapies for Neurodegenerative Diseases. Brain Sci 2024; 14:899. [PMID: 39335395 PMCID: PMC11429837 DOI: 10.3390/brainsci14090899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The role of mitochondria in neurodegenerative diseases is crucial, and recent developments have highlighted its significance in cell therapy. Mitochondrial dysfunction has been implicated in various neurodegenerative disorders, including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, and Huntington's diseases. Understanding the impact of mitochondrial biology on these conditions can provide valuable insights for developing targeted cell therapies. This mini-review refocuses on mitochondria and emphasizes the potential of therapies leveraging mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, stem cell-derived secretions, and extracellular vesicles. Mesenchymal stem cell-mediated mitochondria transfer is highlighted for restoring mitochondrial health in cells with dysfunctional mitochondria. Additionally, attention is paid to gene-editing techniques such as mito-CRISPR, mitoTALENs, mito-ZNFs, and DdCBEs to ensure the safety and efficacy of stem cell treatments. Challenges and future directions are also discussed, including the possible tumorigenic effects of stem cells, off-target effects, disease targeting, immune rejection, and ethical issues.
Collapse
Affiliation(s)
- Adaleiz Ore
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
- Department of Chemical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James M. Angelastro
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
- University of California Medical Investigations of Neurodevelopmental Disorders Institute (MIND Institute), University of California Health, Sacramento, CA 95817, USA
| |
Collapse
|
26
|
Iorio R, Petricca S, Mattei V, Delle Monache S. Horizontal mitochondrial transfer as a novel bioenergetic tool for mesenchymal stromal/stem cells: molecular mechanisms and therapeutic potential in a variety of diseases. J Transl Med 2024; 22:491. [PMID: 38790026 PMCID: PMC11127344 DOI: 10.1186/s12967-024-05047-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/29/2024] [Indexed: 05/26/2024] Open
Abstract
Intercellular mitochondrial transfer (MT) is a newly discovered form of cell-to-cell signalling involving the active incorporation of healthy mitochondria into stressed/injured recipient cells, contributing to the restoration of bioenergetic profile and cell viability, reduction of inflammatory processes and normalisation of calcium dynamics. Recent evidence has shown that MT can occur through multiple cellular structures and mechanisms: tunneling nanotubes (TNTs), via gap junctions (GJs), mediated by extracellular vesicles (EVs) and other mechanisms (cell fusion, mitochondrial extrusion and migrasome-mediated mitocytosis) and in different contexts, such as under physiological (tissue homeostasis and stemness maintenance) and pathological conditions (hypoxia, inflammation and cancer). As Mesenchimal Stromal/ Stem Cells (MSC)-mediated MT has emerged as a critical regulatory and restorative mechanism for cell and tissue regeneration and damage repair in recent years, its potential in stem cell therapy has received increasing attention. In particular, the potential therapeutic role of MSCs has been reported in several articles, suggesting that MSCs can enhance tissue repair after injury via MT and membrane vesicle release. For these reasons, in this review, we will discuss the different mechanisms of MSCs-mediated MT and therapeutic effects on different diseases such as neuronal, ischaemic, vascular and pulmonary diseases. Therefore, understanding the molecular and cellular mechanisms of MT and demonstrating its efficacy could be an important milestone that lays the foundation for future clinical trials.
Collapse
Affiliation(s)
- Roberto Iorio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Sabrina Petricca
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Vincenzo Mattei
- Dipartimento di Scienze della Vita, Della Salute e delle Professioni Sanitarie, Link Campus University, Via del Casale di San Pio V 44, 00165, Rome, Italy.
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy.
| |
Collapse
|
27
|
Lin YH, Lin KL, Wang XW, Lee JJ, Wang FS, Wang PW, Lan MY, Liou CW, Lin TK. Miro1 improves the exogenous engraftment efficiency and therapeutic potential of mitochondria transfer using Wharton's jelly mesenchymal stem cells. Mitochondrion 2024; 76:101856. [PMID: 38408618 DOI: 10.1016/j.mito.2024.101856] [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: 08/24/2023] [Revised: 02/08/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Mitochondria are important for maintaining cellular energy metabolism and regulating cellular senescence. Mitochondrial DNA (mtDNA) encodes subunits of the OXPHOS complexes which are essential for cellular respiration and energy production. Meanwhile, mtDNA variants have been associated with the pathogenesis of neurodegenerative diseases, including MELAS, for which no effective treatment has been developed. To alleviate the pathological conditions involved in mitochondrial disorders, mitochondria transfer therapy has shown promise. Wharton's jelly mesenchymal stem cells (WJMSCs) have been identified as suitable mitochondria donors for mitochondria-defective cells, wherein mitochondrial functions can be rescued. Miro1 participates in mitochondria trafficking by anchoring mitochondria to microtubules. In this study, we identified Miro1 over-expression as a factor that could help to enhance the efficiency of mitochondrial delivery. More specifically, we reveal that Miro1 over-expressed WJMSCs significantly improved intercellular communications, cell proliferation rates, and mitochondrial membrane potential, while restoring mitochondrial bioenergetics in mitochondria-defective fibroblasts. Furthermore, Miro1 over-expressed WJMSCs decreased rates of induced apoptosis and ROS production in MELAS fibroblasts; although, Miro1 over-expression did not rescue mtDNA mutation ratios nor mitochondrial biogenesis. This study presents a potentially novel therapeutic strategy for treating mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS), and other diseases associated with dysfunctional mitochondria, while the pathophysiological relevance of our results should be further verified by animal models and clinical studies.
Collapse
Affiliation(s)
- Yu-Han Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan.
| | - Kai-Lieh Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan.
| | - Xiao-Wen Wang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan.
| | - Jong-Jer Lee
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan.
| | - Feng-Sheng Wang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; Core Laboratory for Phenomics and Diagnostics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan.
| | - Pei-Wen Wang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; Department of Metabolism, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan.
| | - Min-Yu Lan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan; Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan.
| | - Chia-Wei Liou
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan.
| | - Tsu-Kung Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan.
| |
Collapse
|
28
|
Zhang Y, Xu T, Xie J, Wu H, Hu W, Yuan X. MSC-derived mitochondria promote axonal regeneration via Atf3 gene up-regulation by ROS induced DNA double strand breaks at transcription initiation region. Cell Commun Signal 2024; 22:240. [PMID: 38664711 PMCID: PMC11046838 DOI: 10.1186/s12964-024-01617-7] [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: 11/28/2023] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.
Collapse
Affiliation(s)
- Yingchi Zhang
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei, 430030, People's Republic of China
| | - Tao Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei, 430030, People's Republic of China
| | - Jie Xie
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei, 430030, People's Republic of China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei, 430030, People's Republic of China
| | - Weihua Hu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei, 430030, People's Republic of China.
| | - Xuefeng Yuan
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei, 430030, People's Republic of China.
| |
Collapse
|
29
|
Taheri M, Tehrani HA, Dehghani S, Rajabzadeh A, Alibolandi M, Zamani N, Arefian E, Ramezani M. Signaling crosstalk between mesenchymal stem cells and tumor cells: Implications for tumor suppression or progression. Cytokine Growth Factor Rev 2024; 76:30-47. [PMID: 38341337 DOI: 10.1016/j.cytogfr.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stem cells (MSCs) have been extensively used in various therapeutic applications over the last two decades, particularly in regenerative medicine and cancer treatment. MSCs have the ability to differentiate into mesodermal and non-mesodermal lineages, which makes them a popular choice in tissue engineering and regenerative medicine. Studies have shown that MSCs have inherent tumor-suppressive properties and can affect the behavior of multiple cells contributing to tumor development. Additionally, MSCs possess a tumor tropism property and have a hypoimmune nature. The intrinsic features of MSCs along with their potential to undergo genetic manipulation and be loaded with various anticancer therapeutics have motivated researchers to use them in different cancer therapy approaches without considering their complex dynamic biological aspects. However, despite their desirable features, several reports have shown that MSCs possess tumor-supportive properties. These contradictory results signify the sophisticated nature of MSCs and warn against the potential therapeutic applications of MSCs. Therefore, researchers should meticulously consider the biological properties of MSCs in preclinical and clinical studies to avoid any undesirable outcomes. This manuscript reviews preclinical studies on MSCs and cancer from the last two decades, discusses how MSC properties affect tumor progression and explains the mechanisms behind tumor suppressive and supportive functions. It also highlights critical cellular pathways that could be targeted in future studies to improve the safety and effectiveness of MSC-based therapies for cancer treatment. The insights obtained from this study will pave the way for further clinical research on MSCs and development of more effective cancer treatments.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Rajabzadeh
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nina Zamani
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Hazrati A, Malekpour K, Khorramdelazad H, Rajaei S, Hashemi SM. Therapeutic and immunomodulatory potentials of mesenchymal stromal/stem cells and immune checkpoints related molecules. Biomark Res 2024; 12:35. [PMID: 38515166 PMCID: PMC10958918 DOI: 10.1186/s40364-024-00580-2] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are used in many studies due to their therapeutic potential, including their differentiative ability and immunomodulatory properties. These cells perform their therapeutic functions by using various mechanisms, such as the production of anti-inflammatory cytokines, growth factors, direct cell-to-cell contact, extracellular vesicles (EVs) production, and mitochondrial transfer. However, mechanisms related to immune checkpoints (ICPs) and their effect on the immunomodulatory ability of MSCs are less discussed. The main function of ICPs is to prevent the initiation of unwanted responses and to regulate the immune system responses to maintain the homeostasis of these responses. ICPs are produced by various types of immune system regulatory cells, and defects in their expression and function may be associated with excessive responses that can ultimately lead to autoimmunity. Also, by expressing different types of ICPs and their ligands (ICPLs), tumor cells prevent the formation and durability of immune responses, which leads to tumors' immune escape. ICPs and ICPLs can be produced by MSCs and affect immune cell responses both through their secretion into the microenvironment or direct cell-to-cell interaction. Pre-treatment of MSCs in inflammatory conditions leads to an increase in their therapeutic potential. In addition to the effect that inflammatory environments have on the production of anti-inflammatory cytokines by MSCs, they can increase the expression of various types of ICPLs. In this review, we discuss different types of ICPLs and ICPs expressed by MSCs and their effect on their immunomodulatory and therapeutic potential.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Samira Rajaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Liu YB, Hong JR, Jiang N, Jin L, Zhong WJ, Zhang CY, Yang HH, Duan JX, Zhou Y. The Role of Mitochondrial Quality Control in Chronic Obstructive Pulmonary Disease. J Transl Med 2024; 104:100307. [PMID: 38104865 DOI: 10.1016/j.labinv.2023.100307] [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: 06/28/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity, mortality, and health care use worldwide with heterogeneous pathogenesis. Mitochondria, the powerhouses of cells responsible for oxidative phosphorylation and energy production, play essential roles in intracellular material metabolism, natural immunity, and cell death regulation. Therefore, it is crucial to address the urgent need for fine-tuning the regulation of mitochondrial quality to combat COPD effectively. Mitochondrial quality control (MQC) mainly refers to the selective removal of damaged or aging mitochondria and the generation of new mitochondria, which involves mitochondrial biogenesis, mitochondrial dynamics, mitophagy, etc. Mounting evidence suggests that mitochondrial dysfunction is a crucial contributor to the development and progression of COPD. This article mainly reviews the effects of MQC on COPD as well as their specific regulatory mechanisms. Finally, the therapeutic approaches of COPD via MQC are also illustrated.
Collapse
Affiliation(s)
- Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Nan Jiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jia-Xi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
32
|
Han H, Chen BT, Liu Y, Wang Y, Xing L, Wang H, Zhou TJ, Jiang HL. Engineered stem cell-based strategy: A new paradigm of next-generation stem cell product in regenerative medicine. J Control Release 2024; 365:981-1003. [PMID: 38123072 DOI: 10.1016/j.jconrel.2023.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023]
Abstract
Stem cells have garnered significant attention in regenerative medicine owing to their abilities of multi-directional differentiation and self-renewal. Despite these encouraging results, the market for stem cell products yields limited, which is largely due to the challenges faced to the safety and viability of stem cells in vivo. Besides, the fate of cells re-infusion into the body unknown is also a major obstacle to stem cell therapy. Actually, both the functional protection and the fate tracking of stem cells are essential in tissue homeostasis, repair, and regeneration. Recent studies have utilized cell engineering techniques to modify stem cells for enhancing their treatment efficiency or imparting them with novel biological capabilities, in which advances demonstrate the immense potential of engineered cell therapy. In this review, we proposed that the "engineered stem cells" are expected to represent the next generation of stem cell therapies and reviewed recent progress in this area. We also discussed potential applications of engineered stem cells and highlighted the most common challenges that must be addressed. Overall, this review has important guiding significance for the future design of new paradigms of stem cell products to improve their therapeutic efficacy.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Hui Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
33
|
Huang RL, Li Q, Ma JX, Atala A, Zhang Y. Body fluid-derived stem cells - an untapped stem cell source in genitourinary regeneration. Nat Rev Urol 2023; 20:739-761. [PMID: 37414959 PMCID: PMC11639537 DOI: 10.1038/s41585-023-00787-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/08/2023]
Abstract
Somatic stem cells have been obtained from solid organs and tissues, including the bone marrow, placenta, corneal stroma, periosteum, adipose tissue, dental pulp and skeletal muscle. These solid tissue-derived stem cells are often used for tissue repair, disease modelling and new drug development. In the past two decades, stem cells have also been identified in various body fluids, including urine, peripheral blood, umbilical cord blood, amniotic fluid, synovial fluid, breastmilk and menstrual blood. These body fluid-derived stem cells (BFSCs) have stemness properties comparable to those of other adult stem cells and, similarly to tissue-derived stem cells, show cell surface markers, multi-differentiation potential and immunomodulatory effects. However, BFSCs are more easily accessible through non-invasive or minimally invasive approaches than solid tissue-derived stem cells and can be isolated without enzymatic tissue digestion. Additionally, BFSCs have shown good versatility in repairing genitourinary abnormalities in preclinical models through direct differentiation or paracrine mechanisms such as pro-angiogenic, anti-apoptotic, antifibrotic, anti-oxidant and anti-inflammatory effects. However, optimization of protocols is needed to improve the efficacy and safety of BFSC therapy before therapeutic translation.
Collapse
Affiliation(s)
- Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
34
|
Hazrati A, Malekpour K, Mirsanei Z, Khosrojerdi A, Rahmani-Kukia N, Heidari N, Abbasi A, Soudi S. Cancer-associated mesenchymal stem/stromal cells: role in progression and potential targets for therapeutic approaches. Front Immunol 2023; 14:1280601. [PMID: 38022534 PMCID: PMC10655012 DOI: 10.3389/fimmu.2023.1280601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Malignancies contain a relatively small number of Mesenchymal stem/stromal cells (MSCs), constituting a crucial tumor microenvironment (TME) component. These cells comprise approximately 0.01-5% of the total TME cell population. MSC differentiation potential and their interaction with the tumor environment enable these cells to affect tumor cells' growth, immune evasion, metastasis, drug resistance, and angiogenesis. This type of MSC, known as cancer-associated mesenchymal stem/stromal cells (CA-MSCs (interacts with tumor/non-tumor cells in the TME and affects their function by producing cytokines, chemokines, and various growth factors to facilitate tumor cell migration, survival, proliferation, and tumor progression. Considering that the effect of different cells on each other in the TME is a multi-faceted relationship, it is essential to discover the role of these relationships for targeting in tumor therapy. Due to the immunomodulatory role and the tissue repair characteristic of MSCs, these cells can help tumor growth from different aspects. CA-MSCs indirectly suppress antitumor immune response through several mechanisms, including decreasing dendritic cells (DCs) antigen presentation potential, disrupting natural killer (NK) cell differentiation, inducing immunoinhibitory subsets like tumor-associated macrophages (TAMs) and Treg cells, and immune checkpoint expression to reduce effector T cell antitumor responses. Therefore, if these cells can be targeted for treatment so that their population decreases, we can hope for the treatment and improvement of the tumor conditions. Also, various studies show that CA-MSCs in the TME can affect other vital aspects of a tumor, including cell proliferation, drug resistance, angiogenesis, and tumor cell invasion and metastasis. In this review article, we will discuss in detail some of the mechanisms by which CA-MSCs suppress the innate and adaptive immune systems and other mechanisms related to tumor progression.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezou Khosrojerdi
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nasim Rahmani-Kukia
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Heidari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
35
|
Mukkala AN, Jerkic M, Khan Z, Szaszi K, Kapus A, Rotstein O. Therapeutic Effects of Mesenchymal Stromal Cells Require Mitochondrial Transfer and Quality Control. Int J Mol Sci 2023; 24:15788. [PMID: 37958771 PMCID: PMC10647450 DOI: 10.3390/ijms242115788] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Due to their beneficial effects in an array of diseases, Mesenchymal Stromal Cells (MSCs) have been the focus of intense preclinical research and clinical implementation for decades. MSCs have multilineage differentiation capacity, support hematopoiesis, secrete pro-regenerative factors and exert immunoregulatory functions promoting homeostasis and the resolution of injury/inflammation. The main effects of MSCs include modulation of immune cells (macrophages, neutrophils, and lymphocytes), secretion of antimicrobial peptides, and transfer of mitochondria (Mt) to injured cells. These actions can be enhanced by priming (i.e., licensing) MSCs prior to exposure to deleterious microenvironments. Preclinical evidence suggests that MSCs can exert therapeutic effects in a variety of pathological states, including cardiac, respiratory, hepatic, renal, and neurological diseases. One of the key emerging beneficial actions of MSCs is the improvement of mitochondrial functions in the injured tissues by enhancing mitochondrial quality control (MQC). Recent advances in the understanding of cellular MQC, including mitochondrial biogenesis, mitophagy, fission, and fusion, helped uncover how MSCs enhance these processes. Specifically, MSCs have been suggested to regulate peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC1α)-dependent biogenesis, Parkin-dependent mitophagy, and Mitofusins (Mfn1/2) or Dynamin Related Protein-1 (Drp1)-mediated fission/fusion. In addition, previous studies also verified mitochondrial transfer from MSCs through tunneling nanotubes and via microvesicular transport. Combined, these effects improve mitochondrial functions, thereby contributing to the resolution of injury and inflammation. Thus, uncovering how MSCs affect MQC opens new therapeutic avenues for organ injury, and the transplantation of MSC-derived mitochondria to injured tissues might represent an attractive new therapeutic approach.
Collapse
Affiliation(s)
- Avinash Naraiah Mukkala
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mirjana Jerkic
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
| | - Zahra Khan
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Katalin Szaszi
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Andras Kapus
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ori Rotstein
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
36
|
Gao Y, Xu X, Zhang X. Targeting different phenotypes of macrophages: A potential strategy for natural products to treat inflammatory bone and joint diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154952. [PMID: 37506402 DOI: 10.1016/j.phymed.2023.154952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Macrophages, a key class of immune cells, have a dual role in inflammatory responses, switching between anti-inflammatory M2 and pro-inflammatory M1 subtypes depending on the specific environment. Greater numbers of M1 macrophages correlate with increased production of inflammatory chemicals, decreased osteogenic potential, and eventually bone and joint disorders. Therefore, reversing M1 macrophages polarization is advantageous for lowering inflammatory factors. To better treat inflammatory bone disorders in the future, it may be helpful to gain insight into the specific mechanisms and natural products that modulate macrophage polarization. OBJECTIVE This review examines the impact of programmed cell death and different cells in the bone microenvironment on macrophage polarization, as well as the effects of natural products on the various phenotypes of macrophages, in order to suggest some possibilities for the treatment of inflammatory osteoarthritic disorders. METHODS Using 'macrophage polarization,' 'M1 macrophage' 'M2 macrophage' 'osteoporosis,' 'osteonecrosis of femoral head,' 'osteolysis,' 'gouty arthritis,' 'collagen-induced arthritis,' 'freund's adjuvant-induced arthritis,' 'adjuvant arthritis,' and 'rheumatoid arthritis' as search terms, the relevant literature was searched using the PubMed, the Cochrane Library and Web of Science databases. RESULTS Targeting macrophages through different signaling pathways has become a key mechanism for the treatment of inflammatory bone and joint diseases, including HIF-1α, NF-κB, AKT/mTOR, JAK1/2-STAT1, NF-κB, JNK, ERK, p-38α/β, p38/MAPK, PI3K/AKT, AMPK, AMPK/Sirt1, STAT TLR4/NF-κB, TLR4/NLRP3, NAMPT pathway, as well as the programmed cell death autophagy, pyroptosis and ERS. CONCLUSION As a result of a search of databases, we have summarized the available experimental and clinical evidence supporting herbal products as potential treatment agents for inflammatory osteoarthropathy. In this paper, we outline the various modulatory effects of natural substances targeting macrophages in various diseases, which may provide insight into drug options and directions for future clinical trials. In spite of this, more mechanistic studies on natural substances, as well as pharmacological, toxicological, and clinical studies are required.
Collapse
Affiliation(s)
- Yuhe Gao
- Graduate School, Heilongjiang University of Chinese Medicine, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang 150040, China
| | - Xilin Xu
- The Third Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150000, China.
| | - Xiaofeng Zhang
- Teaching and Research Section of Orthopedics and Traumatology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150000, China.
| |
Collapse
|
37
|
Fang X, Lan H, Jin K, Qian J. Pancreatic cancer and exosomes: role in progression, diagnosis, monitoring, and treatment. Front Oncol 2023; 13:1149551. [PMID: 37287924 PMCID: PMC10242099 DOI: 10.3389/fonc.2023.1149551] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 05/05/2023] [Indexed: 06/09/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most dangerous diseases that threaten human life, and investigating the details affecting its progression or regression is particularly important. Exosomes are one of the derivatives produced from different cells, including tumor cells and other cells such as Tregs, M2 macrophages, and MDSCs, and can help tumor growth. These exosomes perform their actions by affecting the cells in the tumor microenvironment, such as pancreatic stellate cells (PSCs) that produce extracellular matrix (ECM) components and immune cells that are responsible for killing tumor cells. It has also been shown that pancreatic cancer cell (PCC)-derived exosomes at different stages carry molecules. Checking the presence of these molecules in the blood and other body fluids can help us in the early stage diagnosis and monitoring of PC. However, immune system cell-derived exosomes (IEXs) and mesenchymal stem cell (MSC)-derived exosomes can contribute to PC treatment. Immune cells produce exosomes as part of the mechanisms involved in the immune surveillance and tumor cell-killing phenomenon. Exosomes can be modified in such a way that their antitumor properties are enhanced. One of these methods is drug loading in exosomes, which can significantly increase the effectiveness of chemotherapy drugs. In general, exosomes form a complex intercellular communication network that plays a role in developing, progressing, diagnosing, monitoring, and treating pancreatic cancer.
Collapse
Affiliation(s)
- Xingliang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
38
|
Kadri N, Amu S, Iacobaeus E, Boberg E, Le Blanc K. Current perspectives on mesenchymal stromal cell therapy for graft versus host disease. Cell Mol Immunol 2023; 20:613-625. [PMID: 37165014 DOI: 10.1038/s41423-023-01022-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/07/2023] [Indexed: 05/12/2023] Open
Abstract
Graft versus host disease (GvHD) is the clinical condition in which bone marrow-derived mesenchymal stromal cells (MSCs) have been most frequently studied. In this review, we summarize the experience from clinical trials that have paved the way to translation. While MSC-based therapy has shown an exceptional safety profile, identifying potency assays and disease biomarkers that reliably predict the capacity of a specific MSC batch to alleviate GvHD has been difficult. As GvHD diagnosis and staging are based solely on clinical criteria, individual patients recruited in the same clinical trial may have vastly different underlying biology, obscuring trial outcomes and making it difficult to determine the benefit of MSCs in subgroups of patients. An accumulating body of evidence indicates the importance of considering not only the cell product but also patient-specific biomarkers and/or immune characteristics in determining MSC responsiveness. A mode of action where intravascular MSC destruction is followed by monocyte-efferocytosis-mediated skewing of the immune repertoire in a permissive inflammatory environment would both explain why cell engraftment is irrelevant for MSC efficacy and stress the importance of biologic differences between responding and nonresponding patients. We recommend a combined analysis of clinical outcomes and both biomarkers of disease activity and MSC potency assays to identify patients with GvHD who are likely to benefit from MSC therapy.
Collapse
Affiliation(s)
- Nadir Kadri
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sylvie Amu
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ellen Iacobaeus
- Department of Clinical Neuroscience, Division of Neurology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Erik Boberg
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Haematology, Karolinska University Hospital, Stockholm, Sweden
| | - Katarina Le Blanc
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Department of Cell Therapies and Allogeneic Stem Cell Transplantation Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
39
|
Malekpour K, Hazrati A, Soudi S, Roshangar L, Pourfathollah AA, Ahmadi M. Combinational administration of mesenchymal stem cell-derived exosomes and metformin reduces inflammatory responses in an in vitro model of insulin resistance in HepG2 cells. Heliyon 2023; 9:e15489. [PMID: 37153436 PMCID: PMC10160701 DOI: 10.1016/j.heliyon.2023.e15489] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetes is a highly common metabolic disorder in advanced societies. One of the causes of diabetes is insulin resistance, which is associated with a loss of sensitivity to insulin-sensitive cells. Insulin resistance develops in the body of a person prone to diabetes many years before diabetes development. Insulin resistance is associated with complications such as hyperglycemia, hyperlipidemia, and compensatory hyperinsulinemia and causes liver inflammation, which, if left untreated, can lead to cirrhosis, fibrosis, and even liver cancer. Metformin is the first line of treatment for patients with diabetes, which lowers blood sugar and increases insulin sensitivity by inhibiting gluconeogenesis in liver cells. The use of metformin has side effects, including a metallic taste in the mouth, vomiting, nausea, diarrhea, and upset stomach. For this reason, other treatments, along with metformin, are being developed. Considering the anti-inflammatory role of mesenchymal stem cells (MSCs) derived exosomes, their use seems to help improve liver tissue function and prevent damage caused by inflammation. This study investigated the anti-inflammatory effect of Wharton's jelly MSCs derived exosomes in combination with metformin in the HepG2 cells insulin resistance model induced by high glucose. This study showed that MSCs derived exosomes as an anti-inflammatory agent in combination with metformin could increase the therapeutic efficacy of metformin without needing to change metformin doses by decreasing inflammatory cytokines production, including IL-1, IL-6, and TNF-α and apoptosis in HepG2 cells.
Collapse
Affiliation(s)
- Kosar Malekpour
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Corresponding author.
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Corresponding author.
| |
Collapse
|