1
|
Wang J, Meng S, Chen Y, Wang H, Hu W, Liu S, Huang L, Xu J, Li Q, Wu X, Huang W, Huang Y. MSC-mediated mitochondrial transfer promotes metabolic reprograming in endothelial cells and vascular regeneration in ARDS. Redox Rep 2025; 30:2474897. [PMID: 40082392 PMCID: PMC11912292 DOI: 10.1080/13510002.2025.2474897] [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] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are a potential therapy for acute respiratory distress syndrome (ARDS), but their mechanisms in repairing mitochondrial damage in ARDS endothelial cells remain unclear. METHODS We first examined MSCs' mitochondrial transfer ability and mechanisms to mouse pulmonary microvascular endothelial cells (MPMECs) in ARDS. Then, we investigated how MSC-mediated mitochondrial transfer affects the repair of endothelial damage. Finally, we elucidated the mechanisms by which MSC-mediated mitochondrial transfer promotes vascular regeneration. RESULTS Compared to mitochondrial-damaged MSCs, normal MSCs showed a significantly higher mitochondrial transfer rate to MPMECs, with increases of 41.68% in vitro (P < 0.0001) and 10.50% in vivo (P = 0.0005). Furthermore, MSC-mediated mitochondrial transfer significantly reduced reactive oxygen species (P < 0.05) and promoted proliferation (P < 0.0001) in MPMECs. Finally, MSC-mediated mitochondrial transfer significantly increased the activity of the tricarboxylic acid (TCA) cycle (MD of CS mRNA: 23.76, P = 0.032), and further enhanced fatty acid synthesis (MD of FAS mRNA: 6.67, P = 0.0001), leading to a 6.7-fold increase in vascular endothelial growth factor release from MPMECs and promoted vascular regeneration in ARDS. CONCLUSION MSC-mediated mitochondrial transfer to MPMECs activates the TCA cycle and fatty acid synthesis, promoting endothelial proliferation and pro-angiogenic factor release, thereby enhancing vascular regeneration in ARDS.
Collapse
Affiliation(s)
- Jinlong Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Shanshan Meng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Yixuan Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Haofei Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Wenhan Hu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Shuai Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Lili Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Jingyuan Xu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Qing Li
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Xiaojing Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Wei Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| | - Yingzi Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
Artusa V, De Luca L, Clerici M, Trabattoni D. Connecting the dots: Mitochondrial transfer in immunity, inflammation, and cancer. Immunol Lett 2025; 274:106992. [PMID: 40054017 DOI: 10.1016/j.imlet.2025.106992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/15/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025]
Abstract
Mitochondria are more than mere energy generators; they are multifaceted organelles that integrate metabolic, signalling, and immune functions, making them indispensable players in maintaining cellular and systemic health. Mitochondrial transfer has recently garnered attention due to its potential role in several physiological and pathological processes. This process involves multiple mechanisms by which mitochondria, along with mitochondrial DNA and other components, are exchanged between cells. In this review, we examine the critical roles of mitochondrial transfer in health and disease, focusing on its impact on immune cell function, the resolution of inflammation, tissue repair, and regeneration. Additionally, we explore its implications in viral infections and cancer progression. We also provide insights into emerging therapeutic applications, emphasizing its potential to address unmet clinical needs.
Collapse
Affiliation(s)
- Valentina Artusa
- Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157 Milan, Italy.
| | - Lara De Luca
- Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157 Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 12, 20122, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 12, 20122, Milan, Italy; IRCCS Fondazione Don Carlo Gnocchi ONLUS, Via Capecelatro 66, 20148 Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157 Milan, Italy.
| |
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
|
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
|
5
|
Castillo-Galán S, Grünenwald F, Hidalgo Y, Cárdenas JC, Cadiz MI, Alcayaga-Miranda F, Khoury M, Cuenca J. Mitochondrial Antiviral Signaling Protein Activation by Retinoic Acid-Inducible Gene I Agonist Triggers Potent Antiviral Defense in Umbilical Cord Mesenchymal Stromal Cells Without Compromising Mitochondrial Function. Int J Mol Sci 2025; 26:4686. [PMID: 40429828 PMCID: PMC12111392 DOI: 10.3390/ijms26104686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Mesenchymal stromal cells (MSCs) represent a promising therapeutic approach in viral infection management. However, their interaction with viruses remains poorly understood. MSCs can support antiviral immune responses and act as viral reservoirs, potentially compromising their therapeutic potential. Innate immune system recognition of viral pathogens involves pattern recognition receptors (PRRs), including RIG-I-like receptors (RLRs), which activate mitochondrial antiviral signaling protein (MAVS). MAVS triggers antiviral pathways like IRF3 and NF-κB, leading to interferon (IFN) production and pro-inflammatory responses. This study explores the antiviral response in umbilical cord-derived MSCs (UC-MSCs) through targeted stimulation with influenza A virus-derived 5'triphosphate-RNA (3p-hpRNA), a RIG-I agonist. By investigating MAVS activation, we provide mechanistic insights into the immune response at the molecular level. Our findings reveal that 3p-hpRNA stimulation triggers immune activation of the IRF3 and NF-κB pathways through MAVS. Subsequently, this leads to the induction of type I and III IFNs, IFN-stimulated genes (ISGs), and pro-inflammatory cytokines. Critically, this immune activation occurs without compromising mitochondrial integrity. UC-MSCs retain their capacity for mitochondrial transfer to recipient cells. These results highlight the adaptability of UC-MSCs, offering a nuanced understanding of immune responses balancing activation with metabolic integrity. Finally, our research provides mechanistic evidence for MSC-based interventions against viral infections.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7550000, Chile; (S.C.-G.); (F.G.); (Y.H.); (F.A.-M.); (M.K.)
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7550000, Chile
| | - Felipe Grünenwald
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7550000, Chile; (S.C.-G.); (F.G.); (Y.H.); (F.A.-M.); (M.K.)
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7550000, Chile
| | - Yessia Hidalgo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7550000, Chile; (S.C.-G.); (F.G.); (Y.H.); (F.A.-M.); (M.K.)
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7550000, Chile
| | - J César Cárdenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 750000, Chile;
- Geroscience Center for Brain Health and Metabolism, Santiago 7750000, Chile
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93101, USA
| | - Maria Ignacia Cadiz
- Cells for Cells, Santiago 7550000, Chile;
- Consorcio REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago 8330024, Chile
| | - Francisca Alcayaga-Miranda
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7550000, Chile; (S.C.-G.); (F.G.); (Y.H.); (F.A.-M.); (M.K.)
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7550000, Chile
- Cells for Cells, Santiago 7550000, Chile;
- Consorcio REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago 8330024, Chile
| | - Maroun Khoury
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7550000, Chile; (S.C.-G.); (F.G.); (Y.H.); (F.A.-M.); (M.K.)
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7550000, Chile
- Cells for Cells, Santiago 7550000, Chile;
- Consorcio REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago 8330024, Chile
| | - Jimena Cuenca
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Santiago 7550000, Chile; (S.C.-G.); (F.G.); (Y.H.); (F.A.-M.); (M.K.)
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago 7550000, Chile
- Cells for Cells, Santiago 7550000, Chile;
- Consorcio REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago 8330024, Chile
| |
Collapse
|
6
|
Im GB, Melero-Martin JM. Mitochondrial transfer in endothelial cells and vascular health. Trends Cell Biol 2025:S0962-8924(25)00105-9. [PMID: 40368738 DOI: 10.1016/j.tcb.2025.04.004] [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: 01/18/2025] [Revised: 03/21/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025]
Abstract
Mitochondria play a vital role in cellular energy metabolism and vascular health, with their function directly influencing endothelial cell (EC) bioenergetics and integrity. Mitochondrial transfer has emerged as a key mechanism of intercellular communication, impacting angiogenesis, tissue repair, and cellular homeostasis. This review highlights recent findings on mitochondrial transfer, including natural mechanisms - such as tunneling nanotubes (TNTs) and extracellular vesicles (EVs) - and artificial approaches like mitochondrial transplantation. These processes enhance EC function and support vascularization under pathological conditions, including ischemia. While early clinical trials demonstrate therapeutic potential, challenges such as mitochondrial instability and scaling host-derived mitochondria persist. Continued research is essential to optimize mitochondrial transfer and advance its application as a therapeutic strategy for restoring vascular health.
Collapse
Affiliation(s)
- Gwang-Bum Im
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
7
|
Li B, Li B, Qiao X, Meng W, Xie Y, Gong J, Fan Y, Zhao Z, Li L. Targeting mitochondrial transfer as a promising therapeutic strategy. Trends Mol Med 2025:S1471-4914(25)00089-9. [PMID: 40335384 DOI: 10.1016/j.molmed.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/17/2025] [Accepted: 04/04/2025] [Indexed: 05/09/2025]
Abstract
Despite the primary impression of mitochondria as energy factories, these organelles are increasingly recognized for their multifaceted roles beyond energy production. Intriguingly, mitochondria can transfer between cells, influencing physiological and pathological processes through intercellular trafficking termed 'mitochondrial transfer.' This phenomenon is important in maintaining metabolic homeostasis, enhancing tissue regeneration, exacerbating cancer progression, and facilitating immune modulation, depending on the cell type and microenvironment. Recently, mitochondrial transfer has emerged as a promising therapeutic target for tissue repair and antitumor therapy. Here, we summarize and critically review recent advances in this field. We aim to provide an updated overview of the mechanisms and potential therapeutic avenues associated with mitochondrial transfer in various diseases from the perspective of different donor cells.
Collapse
Affiliation(s)
- Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Bingzhi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xianghe Qiao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China
| | - Wanrong Meng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuhang Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiajing Gong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Araya-Sapag MJ, Lara-Barba E, García-Guerrero C, Herrera-Luna Y, Flores-Elías Y, Bustamante-Barrientos FA, Albornoz GG, Contreras-Fuentes C, Yantén-Fuentes L, Luque-Campos N, Vega-Letter AM, Toledo J, Luz-Crawford P. New mesenchymal stem/stromal cell-based strategies for osteoarthritis treatment: targeting macrophage-mediated inflammation to restore joint homeostasis. J Mol Med (Berl) 2025:10.1007/s00109-025-02547-8. [PMID: 40272537 DOI: 10.1007/s00109-025-02547-8] [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: 05/27/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Macrophages are pivotal in osteoarthritis (OA) pathogenesis, as their dysregulated polarization can contribute to chronic inflammatory processes. This review explores the molecular and metabolic mechanisms that influence macrophage polarization and identifies potential strategies for OA treatment. Currently, non-surgical treatments for OA focus only on symptom management, and their efficacy is limited; thus, mesenchymal stem/stromal cells (MSCs) have gained attention for their anti-inflammatory and immunomodulatory capabilities. Emerging evidence suggests that small extracellular vesicles (sEVs) derived from MSCs can modulate macrophage function, thus offering potential therapeutic benefits in OA. Additionally, the transfer of mitochondria from MSCs to macrophages has shown promise in enhancing mitochondrial functionality and steering macrophages toward an anti-inflammatory M2-like phenotype. While further research is needed to confirm these findings, MSC-based strategies, including the use of sEVs and mitochondrial transfer, hold great promise for the treatment of OA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- María Jesús Araya-Sapag
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana Lara-Barba
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García-Guerrero
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yesenia Flores-Elías
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Guillermo G Albornoz
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Consuelo Contreras-Fuentes
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile.
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile.
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| |
Collapse
|
9
|
Gao C, Dai Y, Spezza PA, Boasiako P, Tang A, Rasquinha G, Zhong H, Shao B, Liu Y, Shi PA, Lobo CA, An X, Guo A, Mitchell WB, Manwani D, Yazdanbakhsh K, Mendelson A. Megakaryocytes transfer mitochondria to bone marrow mesenchymal stromal cells to lower platelet activation. J Clin Invest 2025; 135:e189801. [PMID: 40014405 PMCID: PMC11996913 DOI: 10.1172/jci189801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
Newly produced platelets acquire a low activation state, but whether the megakaryocyte plays a role in this outcome has not been fully uncovered. Mesenchymal stem cells (MSCs) were previously shown to promote platelet production and lower platelet activation. We found that healthy megakaryocytes transfer mitochondria to MSCs, which is mediated by connexin 43 (Cx43) gap junctions on MSCs and leads to platelets at a low energetic state with increased LYN activation, characteristic of resting platelets with increased LYN activation, characteristic of resting platelets. On the contrary, MSCs have a limited ability to transfer mitochondria to megakaryocytes. Sickle cell disease (SCD) is characterized by hemolytic anemia and results in heightened platelet activation, contributing to numerous disease complications. Platelets in SCD mice and human samples had a heightened energetic state with increased glycolysis. MSC exposure to heme in SCD led to decreased Cx43 expression and a reduced ability to uptake mitochondria from megakaryocytes. This prevented LYN activation in platelets and contributed to increased platelet activation at steady state. Altogether, our findings demonstrate an effect of hemolysis in the microenvironment leading to increased platelet activation in SCD. These findings have the potential to inspire new therapeutic targets to relieve thrombosis-related complications of SCD and other hemolytic conditions.
Collapse
Affiliation(s)
| | - Yitian Dai
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul A. Spezza
- Laboratory of Stem Cell Biology and Engineering Research
| | - Paul Boasiako
- Laboratory of Stem Cell Biology and Engineering Research
| | - Alice Tang
- Laboratory of Stem Cell Biology and Engineering Research
| | | | | | - Bojing Shao
- Laboratory of Vascular Inflammation and Thrombosis Research
| | | | | | - Cheryl A. Lobo
- Laboratory of Blood Borne Parasites, New York Blood Center, New York, New York, USA
| | | | - Anqi Guo
- Laboratory of Complement Biology
| | - William B. Mitchell
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children’s Hospital at Montefiore, Bronx, New York, USA
| | | | | |
Collapse
|
10
|
Li X, Guan Y, Li C, Cheng H, Bai J, Zhao J, Wang Y, Peng J. Recent advances in mitochondrial transplantation to treat disease. BIOMATERIALS TRANSLATIONAL 2025; 6:4-23. [PMID: 40313574 PMCID: PMC12041809 DOI: 10.12336/biomatertransl.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/20/2024] [Accepted: 09/05/2024] [Indexed: 05/03/2025]
Abstract
Mitochondrial transplantation (MT), an innovative regenerative technique widely used to treat diseases caused by mitochondrial dysfunction, shows great promise for clinical application. This procedure can increase the number of mitochondria and improve the function of damaged mitochondria, resulting in increased adenosine triphosphate levels, decreased reactive oxygen species production, improved Ca2+ buffering capacity, modulated inflammatory response, and reduced apoptosis to protect cells, thus promoting tissue repair. In this review, we describe research advances in MT over the last five years, focusing on its application in treating various diseases, including ischaemic injuries (of the kidney, heart, lung, and liver), neurodegenerative disorders, spinal cord injury, sepsis, diabetes mellitus, stroke, and ultraviolet radiation injuries, as well as in procedures such as organ transplantation, focusing on instances where MT demonstrated good efficacy. We also cover the application of engineered mitochondria and mitochondrial combination therapies and present the latest advances in improving MT efficiency, as well as the current clinical applications and shortcomings of MT, aiming to provide a theoretical foundation for enhanced MT utilisation in the future.
Collapse
Affiliation(s)
- Xiangling Li
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
| | - Chaochao Li
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
| | - Haofeng Cheng
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
- Co-Innovation Centre of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
- Department of Neurosurgery, School of Medicine, Nankai University, Tianjin, China
| | - Jun Bai
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
| | - Jinjuan Zhao
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
| | - Yu Wang
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
- Co-Innovation Centre of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Centre of Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 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
|
Chen S, Han C, Shi Z, Guan X, Cheng L, Wang L, Zou W, Liu J. Umbilical mesenchymal stem cells mitigate T-cell compartments shift and Th17/Treg imbalance in acute ischemic stroke via mitochondrial transfer. Stem Cell Res Ther 2025; 16:134. [PMID: 40075467 PMCID: PMC11905603 DOI: 10.1186/s13287-025-04224-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) initiates secondary injuries that worsen neurological damage and hinder recovery. While peripheral immune responses play a key role in stroke outcomes, clinical results from immunotherapy have been suboptimal, with limited focus on T-cell dynamics. Umbilical mesenchymal stem cells (UMSCs) offer therapeutic potential due to their immunomodulatory properties. They can regulate immune responses and reduce neuroinflammation, potentially enhancing recovery by fostering a pro-regenerative peripheral immune environment. However, the effect of UMSCs on T-cell dynamics in AIS remains underexplored. This study investigates T-cell dynamics following AIS and examines how UMSCs may mitigate immune dysregulation to develop better treatment strategies. METHODS AIS patients (NIHSS scores 0-15) were recruited within 72 h of stroke onset, with peripheral blood samples collected on Day 0 (enrollment) and Day 7. T-cell compartments were identified by flow cytometry, and plasma cytokine levels were quantified using a cytometric bead array (CBA). Mitochondria in UMSCs were labeled with MitoTracker. Peripheral blood mononuclear cells from patients were isolated, treated with lipopolysaccharide (LPS), and cocultured with UMSCs in both direct contact and Transwell systems. Flow cytometry, CBA, RT-qPCR, and immunofluorescence assays were used to detect T-cell compartments, gene expression markers for helper T (Th) cell differentiation, cytokine profiles, mitochondrial transfer, reactive oxygen species (ROS) production, and mitochondrial membrane potential. Additionally, mitochondrial DNA in UMSCs was depleted. The effects of UMSCs and mitochondria-depleted UMSCs on ischemic stroke mice were compared through behavioral assessments and analysis of the peripheral immune microenvironment. RESULTS In AIS, T-cell compartments underwent a phenotypic shift from naïve to effector or memory states, with a specific increase in Th17 cells and a decrease in regulatory T cells, leading to alterations in T-cell-mediated immune functions. In an ex vivo co-culture system, LPS stimulation further amplified these disparities, inducing mitochondrial dysfunction and oxidative stress in T cells. Notably, UMSCs restored mitochondrial function and reversed the shift in T-cell compartments through mitochondrial transfer. Critically, UMSC treatment significantly improved both neurological deficits and peripheral immune disorders in ischemic stroke mice, whereas mitochondria-depleted UMSCs failed to produce this effect. CONCLUSIONS Our comprehensive insights into the key attributes of T-cell compartments in acute ischemic stroke and the immune regulatory mechanisms of UMSCs provide a crucial theoretical foundation for understanding peripheral immune disorders in ischemic stroke and the therapeutic potential of UMSC treatment.
Collapse
Affiliation(s)
- Shuna Chen
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Chao Han
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, People's Republic of China
| | - Zihan Shi
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, People's Republic of China
| | - Xin Guan
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
| | - Liyuan Cheng
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Liang Wang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
- College of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, People's Republic of China
| | - Wei Zou
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China.
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, People's Republic of China.
| |
Collapse
|
13
|
Li M, Wu L, Si H, Wu Y, Liu Y, Zeng Y, Shen B. Engineered mitochondria in diseases: mechanisms, strategies, and applications. Signal Transduct Target Ther 2025; 10:71. [PMID: 40025039 PMCID: PMC11873319 DOI: 10.1038/s41392-024-02081-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/30/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Mitochondrial diseases represent one of the most prevalent and debilitating categories of hereditary disorders, characterized by significant genetic, biological, and clinical heterogeneity, which has driven the development of the field of engineered mitochondria. With the growing recognition of the pathogenic role of damaged mitochondria in aging, oxidative disorders, inflammatory diseases, and cancer, the application of engineered mitochondria has expanded to those non-hereditary contexts (sometimes referred to as mitochondria-related diseases). Due to their unique non-eukaryotic origins and endosymbiotic relationship, mitochondria are considered highly suitable for gene editing and intercellular transplantation, and remarkable progress has been achieved in two promising therapeutic strategies-mitochondrial gene editing and artificial mitochondrial transfer (collectively referred to as engineered mitochondria in this review) over the past two decades. Here, we provide a comprehensive review of the mechanisms and recent advancements in the development of engineered mitochondria for therapeutic applications, alongside a concise summary of potential clinical implications and supporting evidence from preclinical and clinical studies. Additionally, an emerging and potentially feasible approach involves ex vivo mitochondrial editing, followed by selection and transplantation, which holds the potential to overcome limitations such as reduced in vivo operability and the introduction of allogeneic mitochondrial heterogeneity, thereby broadening the applicability of engineered mitochondria.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Limin Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haibo Si
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuangang Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
14
|
Lyu X, Yu Y, Jiang Y, Li Z, Qiao Q. The role of mitochondria transfer in cancer biological behavior, the immune system and therapeutic resistance. J Pharm Anal 2025; 15:101141. [PMID: 40115812 PMCID: PMC11925581 DOI: 10.1016/j.jpha.2024.101141] [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: 06/21/2024] [Revised: 10/14/2024] [Accepted: 11/03/2024] [Indexed: 03/23/2025] Open
Abstract
Mitochondria play a crucial role as organelles, managing several physiological processes such as redox balance, cell metabolism, and energy synthesis. Initially, the assumption was that mitochondria primarily resided in the host cells and could exclusively transmit from oocytes to offspring by a mechanism known as vertical inheritance of mitochondria. Recent scholarly works, however, suggest that certain cell types transmit their mitochondria to other developmental cell types via a mechanism referred to as intercellular or horizontal mitochondrial transfer. This review details the process of which mitochondria are transferred across cells and explains the impact of mitochondrial transfer between cells on the efficacy and functionality of cancer cells in various cancer forms. Specifically, we review the role of mitochondria transfer in regulating cellular metabolism restoration, excess reactive oxygen species (ROS) generation, proliferation, invasion, metastasis, mitophagy activation, mitochondrial DNA (mtDNA) inheritance, immune system modulation and therapeutic resistance in cancer. Additionally, we highlight the possibility of using intercellular mitochondria transfer as a therapeutic approach to treat cancer and enhance the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Xintong Lyu
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Yangyang Yu
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Yuanjun Jiang
- Department of Urology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Zhiyuan Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110001, China
| | - Qiao Qiao
- Department of Radiation Oncology, First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
15
|
Kim H, Woo S, Cho HB, Lee S, Cho CW, Park J, Youn S, So G, Kang S, Hwang S, Kim HJ, Park K. Osteoblast-Derived Mitochondria Formulated with Cationic Liposome Guide Mesenchymal Stem Cells into Osteogenic Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412621. [PMID: 39887937 PMCID: PMC11948037 DOI: 10.1002/advs.202412621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/23/2024] [Indexed: 02/01/2025]
Abstract
While mitochondria are known to be essential for intracellular energy production and overall function, emerging evidence highlights their role in influencing cell behavior through mitochondrial transfer. This phenomenon provides a potential basis for the development of treatment strategies for tissue damage and degeneration. This study aims to evaluate whether mitochondria isolated from osteoblasts can promote osteogenic differentiation in mesenchymal stem cells (MSCs). Mitochondria from MSCs, which primarily utilize glycolysis, are compared with those from MG63 cells, which depend on oxidative phosphorylation. Mitochondria from both cell types are then encapsulated in cationic liposomes and transferred to MSCs, and their impact on differentiation is assessed. Mitochondria delivery from MG63 cells to MSCs grown in both two- and three-dimensional cultures results in increased expression of osteogenic markers, including Runt-related transcription factor 2, Osterix, and Osteopontin, and upregulation of genes involved in Bone morphogenetic protein 2 signaling and calcium import. This is accompanied by increased calcium influx and regulated by the Wnt/β-catenin signaling pathway. Transplantation of spheroids containing MSCs with MG63-derived mitochondria in bone defect animal models improves bone regeneration. The results suggest that delivery of MG63-derived mitochondria effectively guides MSCs toward osteogenesis, paving the way for the development of mitochondria-transplantation therapies.
Collapse
Affiliation(s)
- Hye‐Ryoung Kim
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Seonjeong Woo
- Department of Biomedical ScienceCHA UniversitySeongnam13488Republic of Korea
| | - Hui Bang Cho
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sujeong Lee
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Chae Won Cho
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Ji‐In Park
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Seulki Youn
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Gyuwon So
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sumin Kang
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Sohyun Hwang
- Department of Biomedical ScienceCHA UniversitySeongnam13488Republic of Korea
- Department of PathologyCHA Bundang Medical CenterCHA University School of MedicineSeongnam13497Republic of Korea
| | - Hye Jin Kim
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| | - Keun‐Hong Park
- School of BioconvergenceCHA University6F, CHA Biocomplex, Sampyeong‐Dong, Bundang‐guSeongnam‐si13488Republic of Korea
| |
Collapse
|
16
|
Wang H, Li Y, Li H, Yan X, Jiang Z, Feng L, Hu W, Fan Y, Lin S, Li G. T cell related osteoimmunology in fracture healing: Potential targets for augmenting bone regeneration. J Orthop Translat 2025; 51:82-93. [PMID: 39991456 PMCID: PMC11847249 DOI: 10.1016/j.jot.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 02/25/2025] Open
Abstract
UNLABELLED Last decade has witnessed increasing evidence which highlights the roles of immune cells in bone regeneration. Numerous immune cell types, including macrophages, T cells, and neutrophils are involved in fracture healing by orchestrating a series of events that modulate bone formation and remodeling. In this review, the role of T cell immunity in fracture healing has been summarized, and the modulatory effects of T cell immunity in inflammation, bone formation and remodeling have been highlighted. The review also summarizes the specific roles of different T cell subsets, including CD4+ T cells, CD8+ T cells, regulatory T cells, T helper 17 cells, and γδ T cells in modulating fracture healing. The current therapeutics targeting T cell immunity to enhance fracture healing have also been reviewed, aiming to provide insights from a translational standpoint. Overall, this work discusses recent advances and challenges in the interdisciplinary research field of T cell related osteoimmunology and its implications in fracture healing. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Delayed unions or non-unions of bone fractures remain a challenge in clinical practice. Developing a deep understanding of the roles of immune cells, including T cells, in fracture healing will facilitate the advancement of novel therapeutics of fracture nonunion. This review summarizes the current understanding of different T cell subsets involved in various phases of fracture healing, providing insights for targeting T cells as an alternative strategy to enhance bone regeneration.
Collapse
Affiliation(s)
- Haixing Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yashi Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haoxin Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Yan
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhaowei Jiang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Wenhui Hu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Yinuo Fan
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Brestoff JR, Singh KK, Aquilano K, Becker LB, Berridge MV, Boilard E, Caicedo A, Crewe C, Enríquez JA, Gao J, Gustafsson ÅB, Hayakawa K, Khoury M, Lee YS, Lettieri-Barbato D, Luz-Crawford P, McBride HM, McCully JD, Nakai R, Neuzil J, Picard M, Rabchevsky AG, Rodriguez AM, Sengupta S, Sercel AJ, Suda T, Teitell MA, Thierry AR, Tian R, Walker M, Zheng M. Recommendations for mitochondria transfer and transplantation nomenclature and characterization. Nat Metab 2025; 7:53-67. [PMID: 39820558 DOI: 10.1038/s42255-024-01200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/05/2024] [Indexed: 01/19/2025]
Abstract
Intercellular mitochondria transfer is an evolutionarily conserved process in which one cell delivers some of their mitochondria to another cell in the absence of cell division. This process has diverse functions depending on the cell types involved and physiological or disease context. Although mitochondria transfer was first shown to provide metabolic support to acceptor cells, recent studies have revealed diverse functions of mitochondria transfer, including, but not limited to, the maintenance of mitochondria quality of the donor cell and the regulation of tissue homeostasis and remodelling. Many mitochondria-transfer mechanisms have been described using a variety of names, generating confusion about mitochondria transfer biology. Furthermore, several therapeutic approaches involving mitochondria-transfer biology have emerged, including mitochondria transplantation and cellular engineering using isolated mitochondria. In this Consensus Statement, we define relevant terminology and propose a nomenclature framework to describe mitochondria transfer and transplantation as a foundation for further development by the community as this dynamic field of research continues to evolve.
Collapse
Affiliation(s)
- Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Keshav K Singh
- Department of Genetics, I Heersink School of Medicine, University of Alabama at Birmhingham, Birmingham, AL, USA.
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Lance B Becker
- Department of Emergency Medicine, Northwell Health, Manhassett, NY, USA
- Department of Emergency Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Michael V Berridge
- Department of Cancer Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Eric Boilard
- Département de Microbiologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Québec, Canada
| | - Andrés Caicedo
- Instituto de Investigaciones en Biomedicina and Colegio de Ciencias de la Salud, Escuela de Medicina, Universidad San Francisco de Quito, Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
| | - Clair Crewe
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - José Antonio Enríquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de salud Carlos III (CIBERFES), Madrid, Spain
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Åsa B Gustafsson
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratories, Harvard Medical School, Massachusetts General Hospital East 149-2401, Charlestown, MA, USA
| | - Maroun Khoury
- IMPACT Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Universidad de los Andes, Santiago, Chile
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | | | - Patricia Luz-Crawford
- IMPACT Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Universidad de los Andes, Santiago, Chile
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Heidi M McBride
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - James D McCully
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ritsuko Nakai
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Jiri Neuzil
- School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
- Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Alexander G Rabchevsky
- Department of Physiology & the Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Anne-Marie Rodriguez
- UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), Paris, France
| | | | - Alexander J Sercel
- MitoWorld, National Laboratory for Education Transformation, Oakland, CA, USA
| | - Toshio Suda
- Institute of Hematology, Blood Diseases Hospital, Chinese Academy of Sciences and Peking Union Medical College, Tianjin, China
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, Department of Bioengineering, and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Alain R Thierry
- Institute of Research in Cancerology of Montpellier, INSERM U1194, University of Montpellier, ICM, Institut du Cancer de Montpellier, Montpellier, France
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Melanie Walker
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School of the University of Western Australia, Nedlands, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| |
Collapse
|
18
|
Castillo-Galán S, Parra V, Cuenca J. Unraveling the pathogenesis of viral-induced pulmonary arterial hypertension: Possible new therapeutic avenues with mesenchymal stromal cells and their derivatives. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167519. [PMID: 39332781 DOI: 10.1016/j.bbadis.2024.167519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Pulmonary hypertension (PH) is a severe condition characterized by elevated pressure in the pulmonary artery, where metabolic and mitochondrial dysfunction may contribute to its progression. Within the PH spectrum, pulmonary arterial hypertension (PAH) stands out with its primary pulmonary vasculopathy. PAH's prevalence varies from 0.4 to 1.4 per 100,000 individuals and is associated with diverse conditions, including viral infections such as HIV. Notably, recent observations highlight an increased occurrence of PAH among COVID-19 patients, even in the absence of pre-existing cardiopulmonary disorders. While current treatments offer partial relief, there's a pressing need for innovative therapeutic strategies, among which mesenchymal stromal cells (MSCs) and their derivatives hold promise. This review critically evaluates recent investigations into viral-induced PAH, encompassing pathogens like human immunodeficiency virus, herpesvirus, Cytomegalovirus, Hepatitis B and C viruses, SARS-CoV-2, and Human endogenous retrovirus K (HERKV), with a specific emphasis on mitochondrial dysfunction. Furthermore, we explore the underlying rationale driving novel therapeutic modalities, including MSCs, extracellular vesicles, and mitochondrial interventions, within the framework of PAH management.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Valentina Parra
- Laboratory of Differentiation and Cell Metabolism (D&M), Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; SYSTEMIX Center for Systems Biology, O'Higgins University, Rancagua, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile; Cells for Cells, Santiago, Chile.
| |
Collapse
|
19
|
Zuo B, Li X, Xu D, Zhao L, Yang Y, Luan Y, Zhang B. Targeting mitochondrial transfer: a new horizon in cardiovascular disease treatment. J Transl Med 2024; 22:1160. [PMID: 39741312 PMCID: PMC11687156 DOI: 10.1186/s12967-024-05979-x] [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: 09/29/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality among individuals with noncommunicable diseases worldwide. Obesity is associated with an increased risk of developing cardiovascular disease (CVD). Mitochondria are integral to the cardiovascular system, and it has been reported that mitochondrial transfer is associated with the pathogenesis of multiple CVDs and obesity. This review offers a comprehensive examination of the relevance of mitochondrial transfer to cardiovascular health and disease, emphasizing the critical functions of mitochondria in energy metabolism and signal transduction within the cardiovascular system. This highlights how disruptions in mitochondrial transfer contribute to various CVDs, such as myocardial infarction, cardiomyopathies, and hypertension. Additionally, we provide an overview of the molecular mechanisms governing mitochondrial transfer and its potential implications for CVD treatment. This finding underscores the therapeutic potential of mitochondrial transfer and addresses the various mechanisms and challenges in its implementation. By delving into mitochondrial transfer and its targeted modulation, this review aims to advance our understanding of cardiovascular disease treatment, presenting new insights and potential therapeutic strategies in this evolving field.
Collapse
Affiliation(s)
- Baile Zuo
- Molecular Immunology and Immunotherapy Laboratory, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
- Department of Clinical Laboratory, Heping Branch, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dawei Xu
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Liping Zhao
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Yi Luan
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
| | - Bi Zhang
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China.
| |
Collapse
|
20
|
Yin K, Zhang T, Lu X, Shen Q, Gu K, Huang Y, Li C, Hou J, Li J, Zhang G. Tak1 licenses mitochondrial transfer from astrocytes to POMC neurons to maintain glucose and cholesterol homeostasis. Cell Rep 2024; 43:114983. [PMID: 39565693 DOI: 10.1016/j.celrep.2024.114983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/24/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
It remains incompletely understood how the astrocytes in the mediobasal hypothalamus (MBH) regulate systemic glucose and cholesterol metabolism. Here, we show that MBH astrocytic Tak1 (transforming growth factor β [TGF-β]-activated kinase 1) controls the metabolism of glucose and cholesterol. Tak1 is expressed in MBH astrocytes and activated after a short-term nutritional excess. In chow-fed mice, astrocytic deletion of Tak1 across the brain or its suppression in the MBH impairs glucose tolerance, reduces insulin sensitivity, and results in hypercholesterolemia. Astrocytic Tak1 activation in the MBH alleviates these symptoms in mice fed a high-fat diet (HFD). We show that astrocytic Tak1 modulates the activity of proopiomelanocortin (POMC) neurons and enables the transport of mitochondria from astrocytes to POMC neurons. In astrocytic Tak1 knockout mice, supplementation of CD38, a molecule that is crucial in mitochondrial transfer, restores glucose and cholesterol homeostasis. Overall, these findings highlight an important role of MBH astrocytic Tak1 in glucose and cholesterol metabolism.
Collapse
Affiliation(s)
- Kaili Yin
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tingting Zhang
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiyuan Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qing Shen
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Kaiyue Gu
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Ying Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Chaonan Li
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Jingyi Hou
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Guo Zhang
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China.
| |
Collapse
|
21
|
Ji W, Sun L, Wang D, Zhu W. Mesenchymal stem cells alleviate inflammatory responses through regulation of T-cell subsets. Eur J Pharmacol 2024; 983:176996. [PMID: 39277095 DOI: 10.1016/j.ejphar.2024.176996] [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: 06/22/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Immune-mediated inflammatory disease (IMID) is a complex disorder characterized by excessive immune responses involving T cells and their subsets, leading to direct tissue damage. T cells can be broadly categorized into CD4+ T cells and CD8+ T cells. CD4+ T cells are composed of several subsets, including T helper (Th)1, Th2, Th9, Th17, Th22, follicular helper T cells (Tfhs), and regulatory T cells (Tregs), while effector CD8+ T cells consist mainly of cytotoxic T cells (CTLs). Current therapies for IMID are ineffective, prompting exploration into mesenchymal stem cells (MSCs) as a promising clinical treatment due to their immunomodulatory effects and self-renewal potential. Recent studies have shown that MSCs can suppress T cells through direct cell-to-cell contact or secretion of soluble cytokines. Nevertheless, the precise effects of MSCs on T cell subsets remain inadequately defined. In this review, we summarize the most recent studies that have examined how MSCs modulate one or more effector T-cell subsets and the mechanisms behind these modifications in vitro and several mouse models of clinical inflammation. This also provides theoretical support and novel insights into the efficacy of clinical treatments involving MSCs. However, the efficacy of MSC therapies in clinical models of inflammation varies, showing effective remission in most cases, but also with exacerbation of T-cell-mediated inflammatory damage in some instances.
Collapse
Affiliation(s)
- Weimeng Ji
- Department of Oncology, Affiliated Hospital of Jiangsu University, Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu, 212001, China; School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013,China
| | - Li Sun
- Department of Clinical Laboratory, Affiliated Kunshan Hospital Ofjiangsu University, Suzhou, Jiangsu, 215399, China
| | - Deqiang Wang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu, 212001, China.
| | - Wei Zhu
- Department of Oncology, Affiliated Hospital of Jiangsu University, Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu, 212001, China; School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013,China.
| |
Collapse
|
22
|
Baldwin JG, Heuser-Loy C, Saha T, Schelker RC, Slavkovic-Lukic D, Strieder N, Hernandez-Lopez I, Rana N, Barden M, Mastrogiovanni F, Martín-Santos A, Raimondi A, Brohawn P, Higgs BW, Gebhard C, Kapoor V, Telford WG, Gautam S, Xydia M, Beckhove P, Frischholz S, Schober K, Kontarakis Z, Corn JE, Iannacone M, Inverso D, Rehli M, Fioravanti J, Sengupta S, Gattinoni L. Intercellular nanotube-mediated mitochondrial transfer enhances T cell metabolic fitness and antitumor efficacy. Cell 2024; 187:6614-6630.e21. [PMID: 39276774 PMCID: PMC11623344 DOI: 10.1016/j.cell.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 02/20/2024] [Accepted: 08/14/2024] [Indexed: 09/17/2024]
Abstract
Mitochondrial loss and dysfunction drive T cell exhaustion, representing major barriers to successful T cell-based immunotherapies. Here, we describe an innovative platform to supply exogenous mitochondria to T cells, overcoming these limitations. We found that bone marrow stromal cells establish nanotubular connections with T cells and leverage these intercellular highways to transplant stromal cell mitochondria into CD8+ T cells. Optimal mitochondrial transfer required Talin 2 on both donor and recipient cells. CD8+ T cells with donated mitochondria displayed enhanced mitochondrial respiration and spare respiratory capacity. When transferred into tumor-bearing hosts, these supercharged T cells expanded more robustly, infiltrated the tumor more efficiently, and exhibited fewer signs of exhaustion compared with T cells that did not take up mitochondria. As a result, mitochondria-boosted CD8+ T cells mediated superior antitumor responses, prolonging animal survival. These findings establish intercellular mitochondrial transfer as a prototype of organelle medicine, opening avenues to next-generation cell therapies.
Collapse
Affiliation(s)
- Jeremy G Baldwin
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Christoph Heuser-Loy
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Tanmoy Saha
- Center for Engineered Therapeutics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Roland C Schelker
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Dragana Slavkovic-Lukic
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Nicholas Strieder
- Next Generation Sequencing Core, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | | | - Nisha Rana
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; University of Regensburg, Regensburg, Germany
| | - Markus Barden
- Division of Genetic Immunotherapy, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Fabio Mastrogiovanni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Azucena Martín-Santos
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Andrea Raimondi
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Philip Brohawn
- Translational Science and Experimental Medicine, Early R&I, AstraZeneca, Gaithersburg, MD, USA
| | | | - Claudia Gebhard
- Next Generation Sequencing Core, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Veena Kapoor
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William G Telford
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanjivan Gautam
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maria Xydia
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany; Division of Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Philipp Beckhove
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; University of Regensburg, Regensburg, Germany; Division of Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Sina Frischholz
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Zacharias Kontarakis
- Genome Engineering and Measurement Laboratory (GEML), ETH Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, ETH Zürich, University of Zürich, Zürich 8057, Switzerland
| | - Jacob E Corn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Michael Rehli
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Next Generation Sequencing Core, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Jessica Fioravanti
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shiladitya Sengupta
- Center for Engineered Therapeutics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.
| | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; University of Regensburg, Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
23
|
Singer M, Elsayed AM, Husseiny MI. Regulatory T-cells: The Face-off of the Immune Balance. FRONT BIOSCI-LANDMRK 2024; 29:377. [PMID: 39614434 DOI: 10.31083/j.fbl2911377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, ensuring a balanced immune response. Tregs primarily operate in an antigen-specific fashion, facilitated by their distinct distribution within discrete niches. Tregs have been studied extensively, from their point of origin in the thymus origin to their fate in the periphery or organs. Signals received from antigen-presenting cells (APCs) stimulate Tregs to dampen inflammation. Almost all tumors are characterized by a pathological abundance of immune suppression in their microenvironment. Conversely, the lack thereof proves detrimental to immunological disorders. Achieving a balanced expression of Tregs in relation to other immune compartments is important in establishing an effective and adaptable immune tolerance towards cancer cells and autoantigens. In the context of cancer, it is essential to decrease the frequency of Tregs to overcome tumor suppression. A lower survival rate is associated with the presence of excessive exhausted effector immune cells and an increased frequency of regulatory cells. However, when it comes to treating graft rejection and autoimmune diseases, the focus lies on immune tolerance and the transfer of Tregs. Here, we explore the complex mechanisms that Tregs use in human disease to balance effector immune cells.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Ahmed M Elsayed
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
24
|
Vaillant L, Akhter W, Nakhle J, Simon M, Villalba M, Jorgensen C, Vignais ML, Hernandez J. The role of mitochondrial transfer in the suppression of CD8 + T cell responses by Mesenchymal stem cells. Stem Cell Res Ther 2024; 15:394. [PMID: 39497203 PMCID: PMC11536934 DOI: 10.1186/s13287-024-03980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/04/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND . CD8+ Cytotoxic T lymphocytes play a key role in the pathogenesis of autoimmune diseases and clinical conditions such as graft versus host disease and graft rejection. Mesenchymal Stromal Cells (MSCs) are multipotent cells with tissue repair and immunomodulatory capabilities. Since they are able to suppress multiple pathogenic immune responses, MSCs have been proposed as a cellular therapy for the treatment of immune-mediated diseases. However, the mechanisms underlying their immunosuppressive properties are not yet fully understood. MSCs have the remarkable ability to sense tissue injury and inflammation and respond by donating their own mitochondria to neighboring cells. Whether mitochondrial transfer has any role in the repression of CD8+ responses is unknown. METHODS AND RESULTS . We have utilized CD8+ T cells from Clone 4 TCR transgenic mice that differentiate into effector cells upon activation in vitro and in vivo to address this question. Allogeneic bone marrow derived MSCs, co-cultured with activated Clone 4 CD8+ T cells, decreased their expansion, the production of the effector cytokine IFNγ and their diabetogenic potential in vivo. Notably, we found that during this interaction leading to suppression, MSCs transferred mitochondria to CD8+ T cells as evidenced by FACS and confocal microscopy. Transfer of MSC mitochondria to Clone 4 CD8+ T cells also resulted in decreased expansion and production of IFNγ upon activation. These effects overlapped and were additive with those of prostaglandin E2 secreted by MSCs. Furthermore, preventing mitochondrial transfer in co-cultures diminished the ability of MSCs to inhibit IFNγ production. Finally, we demonstrated that both MSCs and MSC mitochondria downregulated T-bet and Eomes expression, key transcription factors for CTL differentiation, on activated CD8+ T cells. CONCLUSION . In this report we showed that MSCs are able to interact with CD8+ T cells and transfer them their mitochondria. Mitochondrial transfer contributed to the global suppressive effect of MSCs on CD8+ T cell activation by downregulating T-bet and Eomes expression resulting in impaired IFNγ production of activated CD8+ T cells.
Collapse
Affiliation(s)
- Loic Vaillant
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Waseem Akhter
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Jean Nakhle
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
- IGMM, Université de Montpellier, CNRS, Montpellier, France
| | - Matthieu Simon
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Martin Villalba
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
| | - Christian Jorgensen
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- CHU Montpellier, Montpellier, France
| | - Marie-Luce Vignais
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Javier Hernandez
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM U1183, Montpellier, 34295, France.
| |
Collapse
|
25
|
Liu Q, Zhang X, Zhu T, Xu Z, Dong Y, Chen B. Mitochondrial transfer from mesenchymal stem cells: Mechanisms and functions. Mitochondrion 2024; 79:101950. [PMID: 39218052 DOI: 10.1016/j.mito.2024.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/04/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Mesenchymal stem cells based therapy has been used in clinic for almost 20 years and has shown encouraging effects in treating a wide range of diseases. However, the underlying mechanism is far more complicated than it was previously assumed. Mitochondria transfer is one way that recently found to be employed by mesenchymal stem cells to exert its biological effects. As one way of exchanging mitochondrial components, mitochondria transfer determines both mesenchymal stem cells and recipient cell fates. In this review, we describe the factors that contribute to MSCs-MT. Then, the routes and mechanisms of MSCs-MT are summarized to provide a theoretical basis for MSCs therapy. Besides, the advantages and disadvantages of MSCs-MT in clinical application are analyzed.
Collapse
Affiliation(s)
- Qing Liu
- Department of Periodontology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Xiaoxin Zhang
- Central laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Tongxin Zhu
- Department of Periodontology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Zhonghan Xu
- Department of Oral Implantology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Yingchun Dong
- Department of Anesthesiology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China.
| | - Bin Chen
- Department of Periodontology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China.
| |
Collapse
|
26
|
Court AC, Vega-Letter AM, Parra-Crisóstomo E, Velarde F, García C, Ortloff A, Vernal R, Pradenas C, Luz-Crawford P, Khoury M, Figueroa FE. Mitochondrial transfer balances cell redox, energy and metabolic homeostasis in the osteoarthritic chondrocyte preserving cartilage integrity. Theranostics 2024; 14:6471-6486. [PMID: 39479450 PMCID: PMC11519804 DOI: 10.7150/thno.96723] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/22/2024] [Indexed: 11/02/2024] Open
Abstract
Osteoarthrosis (OA) is a leading cause of disability and early mortality, with no disease modifying treatment. Mitochondrial (MT) dysfunction and changes in energy metabolism, leading to oxidative stress and apoptosis, are main drivers of disease. In reaction to stress, mesenchymal stromal/stem cells (MSCs) donate their MT to damaged tissues. Methods: To evaluate the capacity of clinically validated MSCs to spontaneously transfer their MT to human OA chondrocytes (OA-Ch), primary cultured Ch isolated from the articular cartilage of OA patients were co-cultured with MT-labeled MSCs. MT transfer (MitoT) was evidenced by flow cytometry and confocal microscopy of MitoTracker-stained and YFP-tagged MT protein. MT persistence and metabolic analysis on target cells were assessed by direct transfer of MSC-derived MT to OA-Chs (Mitoception), through SNP-qPCR analysis, ATP measurements and Seahorse technology. The effects of MitoT on MT dynamics, oxidative stress and cell viability were gauged by western blot of fusion/fission proteins, confocal image analysis, ROS levels, Annexin V/7AAD and TUNEL assays. Intra-articular injection of MSC-derived MT was tested in a collagenase-induced murine model of OA. Results: Dose-dependent cell-to-cell MitoT from MSCs to cultured OA-Chs was detected starting at 4 hours of co-culture, with increasing MT-fluorescence levels at higher MSC:Ch ratios. PCR analysis confirmed the presence of exogenous MSC-MT within MitoT+ OA-Chs up to 9 days post Mitoception. MitoT from MSCs to OA-Ch restores energetic status, with a higher ATP production and metabolic OXPHOS/Glycolisis ratio. Significant changes in the expression of MT network regulators, increased MFN2 and decreased p-DRP1, reveal that MitoT promotes MT fusion restoring the MT dynamics in the OA-Ch. Additionally, MitoT increases SOD2 transcripts, protein, and activity levels, and reduces ROS levels, confering resistance to oxidative stress and enhancing resistance to apoptosis. Intra-articular injection of MSC-derived MT improves histologic scores and bone density of the affected joints in the OA mouse model, demonstrating a protective effect of MT transplantation on cartilage degradation. Conclusion: The Mitochondria transfer of MSC-derived MT induced reversal of the metabolic dysfunction by restoring the energetic status and mitochondrial dynamics in the OA chondrocyte, while conferring resistance to oxidative stress and apoptosis. Intra-articular injection of MT improved the disease in collagenase-induced OA mouse model. The restoration of the cellular homeostasis and the preclinical benefit of the intra-articular MT treatment offer a new approach for the treatment of OA.
Collapse
Affiliation(s)
- Angela C. Court
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliseo Parra-Crisóstomo
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Francesca Velarde
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Rolando Vernal
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carolina Pradenas
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maroun Khoury
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Fernando E. Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
27
|
Nakai R, Varnum S, Field RL, Shi H, Giwa R, Jia W, Krysa SJ, Cohen EF, Borcherding N, Saneto RP, Tsai RC, Suganuma M, Ohta H, Yokota T, Brestoff JR. Mitochondria transfer-based therapies reduce the morbidity and mortality of Leigh syndrome. Nat Metab 2024; 6:1886-1896. [PMID: 39223312 DOI: 10.1038/s42255-024-01125-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria transfer is a recently described phenomenon in which donor cells deliver mitochondria to acceptor cells1-3. One possible consequence of mitochondria transfer is energetic support of neighbouring cells; for example, exogenous healthy mitochondria can rescue cell-intrinsic defects in mitochondrial metabolism in cultured ρ0 cells or Ndufs4-/- peritoneal macrophages4-7. Exposing haematopoietic stem cells to purified mitochondria before autologous haematopoietic stem cell transplantation allowed for treatment of anaemia in patients with large-scale mitochondrial DNA mutations8,9, and mitochondria transplantation was shown to minimize ischaemic damage to the heart10-12, brain13-15 and limbs16. However, the therapeutic potential of using mitochondria transfer-based therapies to treat inherited mitochondrial diseases is unclear. Here we demonstrate improved morbidity and mortality of the Ndufs4-/- mouse model of Leigh syndrome (LS) in multiple treatment paradigms associated with mitochondria transfer. Transplantation of bone marrow from wild-type mice, which is associated with release of haematopoietic cell-derived extracellular mitochondria into circulation and transfer of mitochondria to host cells in multiple organs, ameliorates LS in mice. Furthermore, administering isolated mitochondria from wild-type mice extends lifespan, improves neurological function and increases energy expenditure of Ndufs4-/- mice, whereas mitochondria from Ndufs4-/- mice did not improve neurological function. Finally, we demonstrate that cross-species administration of human mitochondria to Ndufs4-/- mice also improves LS. These data suggest that mitochondria transfer-related approaches can be harnessed to treat mitochondrial diseases, such as LS.
Collapse
Affiliation(s)
- Ritsuko Nakai
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
- Department of Hematology, Sakai City Medical Center, Sakai, Japan
| | - Stella Varnum
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachael L Field
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Henyun Shi
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Rocky Giwa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wentong Jia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Samantha J Krysa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eva F Cohen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Russell P Saneto
- Neuroscience Institute, Center for Integrated Brain Research, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | | | | | | | - Takafumi Yokota
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan.
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan.
| | - Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
28
|
Bourebaba L, Bourebaba N, Galuppo L, Marycz K. Artificial mitochondrial transplantation (AMT) reverses aging of mesenchymal stromal cells and improves their immunomodulatory properties in LPS-induced synoviocytes inflammation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119806. [PMID: 39098401 DOI: 10.1016/j.bbamcr.2024.119806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Nowadays, regenerative medicine techniques are usually based on the application of mesenchymal stromal cells (MSCs) for the repair or restoration of injured damaged tissues. However, the effectiveness of autologous therapy is limited as therapeutic potential of MSCs declines due to patient's age, health condition and prolonged in vitro cultivation as a result of decreased growth rate. For that reason, there is an urgent need to develop strategies enabling the in vitro rejuvenation of MSCs prior transplantation in order to enhance their in vivo therapeutic efficiency. In presented study, we attempted to mimic the naturally occurring mitochondrial transfer (MT) between neighbouring cells and verify whether artificial MT (AMT) could reverse MSCs aging and improve their biological properties. For that reason, mitochondria were isolated from healthy donor equine adipose-derived stromal cells (ASCs) and transferred into metabolically impaired recipient ASCs derived from equine metabolic syndrome (EMS) affected horses, which were subsequently subjected to various analytical methods in order to verify the cellular and molecular outcomes of the applied AMT. Mitochondria recipient cells were characterized by decreased apoptosis, senescence and endoplasmic reticulum stress while insulin sensitivity was enhanced. Furthermore, we observed increased mitochondrial fragmentation and associated PARKIN protein accumulation, which indicates on the elimination of dysfunctional organelles via mitophagy. AMT further promoted physioxia and regulated autophagy fluxes. Additionally, rejuvenated ASCs displayed an improved anti-inflammatory activity toward LPS-stimulated synoviocytes. The presented findings highlight AMT as a promising alternative and effective method for MSCs rejuvenation, for potential application in autologous therapies in which MSCs properties are being strongly deteriorated due to patients' condition.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375 Wroclaw, Poland.
| | - Nabila Bourebaba
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375 Wroclaw, Poland
| | - Larry Galuppo
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA 95516, United States
| | - Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375 Wroclaw, Poland; Department of Veterinary Medicine and Epidemiology, Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA 95516, United States.
| |
Collapse
|
29
|
Jia L, Li N, van Unen V, Zwaginga JJ, Braun J, Hiemstra PS, Koning F, Khedoe PPSJ, Stolk J. Pulmonary and Systemic Immune Profiles Following Lung Volume Reduction Surgery and Allogeneic Mesenchymal Stromal Cell Treatment in Emphysema. Cells 2024; 13:1636. [PMID: 39404398 PMCID: PMC11476308 DOI: 10.3390/cells13191636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Emphysema in patients with chronic obstructive pulmonary disease (COPD) is characterized by progressive inflammation. Preclinical studies suggest that lung volume reduction surgery (LVRS) and mesenchymal stromal cell (MSC) treatment dampen inflammation. We investigated the effects of bone marrow-derived MSC (BM-MSC) and LVRS on circulating and pulmonary immune cell profiles in emphysema patients using mass cytometry. Blood and resected lung tissue were collected at the first LVRS (L1). Following 6-10 weeks of recovery, patients received a placebo or intravenous administration of 2 × 106 cells/kg bodyweight BM-MSC (n = 5 and n = 9, resp.) in week 3 and 4 before the second LVRS (L2), where blood and lung tissue were collected. Irrespective of BM-MSC or placebo treatment, proportions of circulating lymphocytes including central memory CD4 regulatory, effector memory CD8 and γδ T cells were higher, whereas myeloid cell percentages were lower in L2 compared to L1. In resected lung tissue, proportions of Treg (p = 0.0067) and anti-inflammatory CD163- macrophages (p = 0.0001) were increased in L2 compared to L1, while proportions of pro-inflammatory CD163+ macrophages were decreased (p = 0.0004). There were no effects of BM-MSC treatment on immune profiles in emphysema patients. However, we observed alterations in the circulating and pulmonary immune cells upon LVRS, suggesting the induction of anti-inflammatory responses potentially needed for repair processes.
Collapse
Affiliation(s)
- Li Jia
- Department of Immunology, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands; (L.J.)
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands (J.S.)
| | - Na Li
- Department of Immunology, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands; (L.J.)
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory of Zoonosis Research of the Ministry of Education, Institute of Zoonosis and College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Vincent van Unen
- Department of Immunology, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands; (L.J.)
| | - Jaap-Jan Zwaginga
- Department of Hematology, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Jerry Braun
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2333 Leiden, The Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands (J.S.)
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands; (L.J.)
| | - P. Padmini S. J. Khedoe
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands (J.S.)
| | - Jan Stolk
- Department of Pulmonology, PulmoScience Lab, Leiden University Medical Center (LUMC), 2333 Leiden, The Netherlands (J.S.)
| |
Collapse
|
30
|
Wen F, Yang G, Yu S, Liu H, Liao N, Liu Z. Mesenchymal stem cell therapy for liver transplantation: clinical progress and immunomodulatory properties. Stem Cell Res Ther 2024; 15:320. [PMID: 39334441 PMCID: PMC11438256 DOI: 10.1186/s13287-024-03943-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Although liver transplantation (LT) is an effective strategy for end-stage liver diseases, the shortage of donor organs and the immune rejection hinder its widespread implementation in clinical practice. Mesenchymal stem cells (MSCs) transplantation offers a promising approach for patients undergoing liver transplantation due to their immune regulatory capabilities, hepatic protection properties, and multidirectional differentiation potential. In this review, we summarize the potential applications of MSCs transplantation in various LT scenarios. MSCs transplantation has demonstrated effectiveness in alleviating hepatic ischemia-reperfusion injury, enhancing the viability of liver grafts, preventing acute graft-versus-host disease, and promoting liver regeneration in split LT therapy. We also discuss the clinical progress, and explore the immunomodulatory functions of MSCs in response to both adaptive and innate immune responses. Furthermore, we emphasize the interactions between MSCs and different immune cells, including T cells, B cells, plasma cells, natural killer cells, dendritic cells, Kupffer cells, and neutrophils, to provide new insights into the immunomodulatory properties of MSCs in adoptive cell therapy.
Collapse
Affiliation(s)
- Fuli Wen
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Guokai Yang
- Department of Nephrology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Saihua Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China
| | - Haiyan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China.
| | - Zhengfang Liu
- Department of Traditional Chinese Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China.
| |
Collapse
|
31
|
Court AC, Parra-Crisóstomo E, Castro-Córdova P, Abdo L, Aragão EAA, Lorca R, Figueroa FE, Bonamino MH, Khoury M. Survival advantage of native and engineered T cells is acquired by mitochondrial transfer from mesenchymal stem cells. J Transl Med 2024; 22:868. [PMID: 39334383 PMCID: PMC11429672 DOI: 10.1186/s12967-024-05627-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Apoptosis, a form of programmed cell death, is critical for the development and homeostasis of the immune system. Chimeric antigen receptor T (CAR-T) cell therapy, approved for hematologic cancers, retains several limitations and challenges associated with ex vivo manipulation, including CAR T-cell susceptibility to apoptosis. Therefore, strategies to improve T-cell survival and persistence are required. Mesenchymal stem/stromal cells (MSCs) exhibit immunoregulatory and tissue-restoring potential. We have previously shown that the transfer of umbilical cord MSC (UC-MSC)-derived mitochondrial (MitoT) prompts the genetic reprogramming of CD3+ T cells towards a Treg cell lineage. The potency of T cells plays an important role in effective immunotherapy, underscoring the need for improving their metabolic fitness. In the present work, we evaluate the effect of MitoT on apoptotis of native T lymphocytes and engineered CAR-T cells. METHODS We used a cell-free approach using artificial MitoT (Mitoception) of UC-MSC derived MT to peripheral blood mononuclear cells (PBMCs) followed by RNA-seq analysis of CD3+ MitoTpos and MitoTneg sorted cells. Target cell apoptosis was induced with Staurosporine (STS), and cell viability was evaluated with Annexin V/7AAD and TUNEL assays. Changes in apoptotic regulators were assessed by flow cytometry, western blot, and qRT-PCR. The effect of MitoT on 19BBz CAR T-cell apoptosis in response to electroporation with a non-viral transposon-based vector was assessed with Annexin V/7AAD. RESULTS Gene expression related to apoptosis, cell death and/or responses to different stimuli was modified in CD3+ T cells after Mitoception. CD3+MitoTpos cells were resistant to STS-induced apoptosis compared to MitoTneg cells, showing a decreased percentage in apoptotic T cells as well as in TUNEL+ cells. Additionally, MitoT prevented the STS-induced collapse of the mitochondrial membrane potential (MMP) levels, decreased caspase-3 cleavage, increased BCL2 transcript levels and BCL-2-related BARD1 expression in FACS-sorted CD3+ T cells. Furthermore, UC-MSC-derived MitoT reduced both early and late apoptosis in CAR-T cells following electroporation, and exhibited an increasing trend in cytotoxic activity levels. CONCLUSIONS Artificial MitoT prevents STS-induced apoptosis of human CD3+ T cells by interfering with the caspase pathway. Furthermore, we observed that MitoT confers protection to apoptosis induced by electroporation in MitoTpos CAR T-engineered cells, potentially improving their metabolic fitness and resistance to environmental stress. These results widen the physiological perspective of organelle-based therapies in immune conditions while offering potential avenues to enhance CAR-T treatment outcomes where their viability is compromised.
Collapse
Affiliation(s)
- Angela C Court
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Eliseo Parra-Crisóstomo
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Pablo Castro-Córdova
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Luiza Abdo
- Cell and Gene Therapy Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | | | - Rocío Lorca
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Fernando E Figueroa
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
- Consorcio Regenero and R-MATIS, Chilean Consortium for Regenerative Medicine, and Manufacture of Advanced Therapies for Innovative Science, Santiago, Chile
| | - Martín Hernán Bonamino
- Cell and Gene Therapy Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation, (FIOCRUZ), Rio de Janeiro, Brazil
| | - Maroun Khoury
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cell for Cells, Santiago, Chile.
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile.
- Consorcio Regenero and R-MATIS, Chilean Consortium for Regenerative Medicine, and Manufacture of Advanced Therapies for Innovative Science, Santiago, Chile.
| |
Collapse
|
32
|
Shen M, Zhou L, Fan X, Wu R, Liu S, Deng Q, Zheng Y, Liu J, Yang L. Metabolic Reprogramming of CD4 + T Cells by Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuates Autoimmune Hepatitis Through Mitochondrial Protein Transfer. Int J Nanomedicine 2024; 19:9799-9819. [PMID: 39345912 PMCID: PMC11430536 DOI: 10.2147/ijn.s472086] [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: 06/09/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Background Autoimmune hepatitis (AIH) is a serious liver disease characterized by immune disorders, particularly effector T-cell overactivation. This study aimed to explore the therapeutic effect and underlying mechanism of mesenchymal stem cell-derived extracellular vesicle (MSC-EV) treatment on CD4+ T-cell overactivation and liver injury in AIH. Methods The metabolic changes of CD4+ T cells were assayed in human AIH and mouse hepatitis models. The liver protective effect of MSC-EVs was evaluated by transaminase levels, liver histopathology and inflammation. The effect of MSC-EVs on the metabolic state of CD4+ T cells was also explored. Results Enhanced glycolysis (eg, ~1.5-fold increase in hexokinase 2 levels) was detected in the CD4+ T cells of AIH patient samples and mouse hepatitis models, whereas the inhibition of glycolysis decreased CD4+ T-cell activation (~1.8-fold decrease in CD69 levels) and AIH liver injury (~6-fold decrease in aminotransferase levels). MSC-EV treatment reduced CD4+ T-cell activation (~1.5-fold decrease in CD69 levels) and cytokine release (~5-fold decrease in IFN-γ levels) by reducing glycolysis (~3-fold decrease) while enhancing mitochondrial oxidative phosphorylation (~2-fold increase in maximal respiration) in such cells. The degree of liver damage in AIH mice was ameliorated after MSC-EV treatment (~5-fold decrease in aminotransferase levels). MSC-EVs carried abundant mitochondrial proteins and might transfer them to metabolically reprogram CD4+ T cells, whereas disrupting mitochondrial transfer impaired the therapeutic potency of MSC-EVs in activated CD4+ T cells. Conclusion Disordered glucose metabolism promotes CD4+ T-cell activation and associated inflammatory liver injury in AIH models, which can be reversed by MSC-EV therapy, and this effect is at least partially dependent on EV-mediated mitochondrial protein transfer between cells. This study highlights that MSC-EV therapy may represent a new avenue for treating autoimmune diseases such as AIH.
Collapse
Affiliation(s)
- Mengyi Shen
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Leyu Zhou
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ruiqi Wu
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Shuyun Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qiaoyu Deng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Li Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
33
|
Zhou M, Sun R, Jang J, Martin JG. T cell and airway smooth muscle interaction: a key driver of asthmatic airway inflammation and remodeling. Am J Physiol Lung Cell Mol Physiol 2024; 327:L382-L394. [PMID: 39010821 DOI: 10.1152/ajplung.00121.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Abstract
Cross talk between T cells and airway smooth muscle (ASM) may play a role in modulating asthmatic airway inflammation and remodeling. Infiltrating T cells have been observed within the ASM bundles of asthmatics, and a wide range of direct and indirect interactions between T cells and ASM has been demonstrated using various in vitro and in vivo model systems. Contact-dependent mechanisms such as ligation and activation of cellular adhesion and costimulatory molecules, as well as the formation of lymphocyte-derived membrane conduits, facilitate the adhesion, bidirectional communication, and transfer of materials between T and ASM cells. T cell-derived cytokines, particularly of the Th1, Th2, and Th17 subsets, modulate the secretome, proliferation, and contractility of ASM cells. This review summarizes the mechanisms governing T cell-ASM cross talk in the context of asthma. Understanding the underlying mechanistic basis is important for directing future research and developing therapeutic interventions targeted toward this complex interaction.
Collapse
Affiliation(s)
- Muyang Zhou
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rui Sun
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Joyce Jang
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - James G Martin
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
34
|
Hu W, Deng J, Su Z, Wang H, Lin S. Advances on T cell immunity in bone remodeling and bone regeneration. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:450-459. [PMID: 39183057 PMCID: PMC11375490 DOI: 10.3724/zdxbyxb-2023-0619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 07/29/2024] [Indexed: 08/27/2024]
Abstract
Bone remodeling and bone regeneration are essential for preserving skeletal integrity and maintaining mineral homeostasis. T cells, as key members of adaptive immunity, play a pivotal role in bone remodeling and bone regeneration by producing a range of cytokines and growth factors. In the physiological state, T cells are involved in the maintenance of bone homeostasis through interactions with mesenchymal stem cells, osteoblasts, and osteoclasts. In pathological states, T cells participate in the pathological process of different types of osteoporosis through interaction with estrogen, glucocorticoids, and parathyroid hormone. During fracture healing for post-injury repair, T cells play different roles during the inflammatory hematoma phase, the bone callus formation phase and the bone remodeling phase. Targeting T cells thus emerges as a potential strategy for regulating bone homeostasis. This article reviews the research progress on related mechanisms of T cells immunity involved in bone remodeling and bone regeneration, with a view to providing a scientific basis for targeting T cells to regulate bone remodeling and bone regeneration.
Collapse
Affiliation(s)
- Wenhui Hu
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China.
| | - Jinxia Deng
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Zhanpeng Su
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Haixing Wang
- Department of Orthopedics and Traumatology, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hongkong 999077, China
| | - Sien Lin
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China.
- Department of Orthopedics and Traumatology, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hongkong 999077, China.
| |
Collapse
|
35
|
Qin X, Liu J. Nanoformulations for the diagnosis and treatment of metabolic dysfunction-associated steatohepatitis. Acta Biomater 2024; 184:37-53. [PMID: 38879104 DOI: 10.1016/j.actbio.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive phase of metabolic dysfunction-associated steatotic liver disease (MASLD) that develops into irreversible liver cirrhosis and hepatocellular carcinoma, ultimately necessitating liver transplantation as the sole life-saving option. However, given the drawbacks of liver transplantation, including invasiveness, chronic immunosuppression, and a lack of donor livers, prompt diagnosis and effective treatment are indispensable. Due to the limitations of liver biopsy and conventional imaging modalities in diagnosing MASH, as well as the potential hazards associated with liver-protecting medicines, numerous nanoformulations have been created for MASH theranostics. Particularly, there has been significant study interest in artificial nanoparticles, natural biomaterials, and bionic nanoparticles that exhibit exceptional biocompatibility and bioavailability. In this review, we summarized extracellular vesicles (EVs)-based omics analysis and Fe3O4-based functional magnetic nanoparticles as magnetic resonance imaging (MRI) contrast agents for MASH diagnosis. Additionally, artificial nanoparticles such as organic and inorganic nanoparticles, as well as natural biomaterials such as cells and cell-derived EVs and bionic nanoparticles including cell membrane-coated nanoparticles, have also been reported for MASH treatment owing to their specific targeting and superior therapeutic effect. This review has the potential to stimulate advancements in nanoformulation fabrication techniques. By exploring their compatibility with cell biology, it could lead to the creation of innovative material systems for efficient theragnostic uses for MASH. STATEMENT OF SIGNIFICANCE: People with metabolic dysfunction-associated steatohepatitis (MASH) will progress to fibrosis, cirrhosis, or even liver cancer. It is imperative to establish effective theragnostic techniques to stop MASH from progressing into a lethal condition. In our review, we summarize the advancement of artificial, natural, and bionic nanoparticles applied in MASH theragnosis. Furthermore, the issues that need to be resolved for these cutting-edge techniques are summarized to realize a more significant clinical impact. We forecast the key fields that will advance further as nanotechnology and MASH research progress. Generally, our discovery has significant implications for the advancement of nanoformulation fabrication techniques, and their potential to be compatible with cell biology could lead to the creation of innovative materials systems for effective MASH theragnostic.
Collapse
Affiliation(s)
- Xueying Qin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China
| | - Jingjing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, PR China.
| |
Collapse
|
36
|
Hu H, Li H, Li R, Liu P, Liu H. Re-establishing immune tolerance in multiple sclerosis: focusing on novel mechanisms of mesenchymal stem cell regulation of Th17/Treg balance. J Transl Med 2024; 22:663. [PMID: 39010157 PMCID: PMC11251255 DOI: 10.1186/s12967-024-05450-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
The T-helper 17 (Th17) cell and regulatory T cell (Treg) axis plays a crucial role in the development of multiple sclerosis (MS), which is regarded as an immune imbalance between pro-inflammatory cytokines and the maintenance of immune tolerance. Mesenchymal stem cell (MSC)-mediated therapies have received increasing attention in MS research. In MS and its animal model experimental autoimmune encephalomyelitis, MSC injection was shown to alter the differentiation of CD4+T cells. This alteration occurred by inducing anergy and reduction in the number of Th17 cells, stimulating the polarization of antigen-specific Treg to reverse the imbalance of the Th17/Treg axis, reducing the inflammatory cascade response and demyelination, and restoring an overall state of immune tolerance. In this review, we summarize the mechanisms by which MSCs regulate the balance between Th17 cells and Tregs, including extracellular vesicles, mitochondrial transfer, metabolic reprogramming, and autophagy. We aimed to identify new targets for MS treatment using cellular therapy by analyzing MSC-mediated Th17-to-Treg polarization.
Collapse
Affiliation(s)
- Huiru Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Hui Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ruoyu Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Peidong Liu
- Department of Neurosurgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Translational Medicine Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Hongbo Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Translational Medicine Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
37
|
Li H, Lei Y, Lai X, Huang R, Xiang Y, Zhao Z, Fang Z, Lai T. Comprehensive analysis and identification of subtypes and hub genes of high immune response in lung adenocarcinoma. BMC Pulm Med 2024; 24:324. [PMID: 38965571 PMCID: PMC11225283 DOI: 10.1186/s12890-024-03130-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 06/24/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND The advent of immunotherapy targeting immune checkpoints has conferred significant clinical advantages to patients with lung adenocarcinoma (LUAD); However, only a limited subset of patients exhibit responsiveness to this treatment. Consequently, there is an imperative need to stratify LUAD patients based on their response to immunotherapy and enhance the therapeutic efficacy of these treatments. METHODS The differentially co-expressed genes associated with CD8 + T cells were identified through weighted gene co-expression network analysis (WGCNA) and the Search Tool for the Retrieval of Interacting Genes (STRING) database. These gene signatures facilitated consensus clustering for TCGA-LUAD and GEO cohorts, categorizing them into distinct immune subtypes (C1, C2, C3, and C4). The Tumor Immune Dysfunction and Exclusion (TIDE) model and Immunophenoscore (IPS) analysis were employed to assess the immunotherapy response of these subtypes. Additionally, the impact of inhibitors targeting five hub genes on the interaction between CD8 + T cells and LUAD cells was evaluated using CCK8 and EDU assays. To ascertain the effects of these inhibitors on immune checkpoint genes and the cytotoxicity mediated by CD8 + T cells, flow cytometry, qPCR, and ELISA methods were utilized. RESULTS Among the identified immune subtypes, subtypes C1 and C3 were characterized by an abundance of immune components and enhanced immunogenicity. Notably, both C1 and C3 exhibited higher T cell dysfunction scores and elevated expression of immune checkpoint genes. Multi-cohort analysis of Lung Adenocarcinoma (LUAD) suggested that these subtypes might elicit superior responses to immunotherapy and chemotherapy. In vitro experiments involved co-culturing LUAD cells with CD8 + T cells and implementing the inhibition of five pivotal genes to assess their function. The inhibition of these genes mitigated the immunosuppression on CD8 + T cells, reduced the levels of PD1 and PD-L1, and promoted the secretion of IFN-γ and IL-2. CONCLUSIONS Collectively, this study delineated LUAD into four distinct subtypes and identified five hub genes correlated with CD8 + T cell activity. It lays the groundwork for refining personalized therapy and immunotherapy strategies for patients with LUAD.
Collapse
Affiliation(s)
- Han Li
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Yuting Lei
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xianwen Lai
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Ruina Huang
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Yuanyuan Xiang
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Zhao Zhao
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Zhenfu Fang
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China.
| |
Collapse
|
38
|
Zhang Q, Zeng Z, Wei N, Su Y, Wang J, Ni Q, Wang Y, Yang J, Liu X, Xu H, Wang G, Shan Y, Zhou F. Mesenteric lymph nodes: a critical site for the up-regulatory effect of hUC-MSCs on Treg cells by producing TGF-β1 in colitis treatment. Stem Cell Res Ther 2024; 15:190. [PMID: 38956621 PMCID: PMC11218300 DOI: 10.1186/s13287-024-03809-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: 02/27/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) demonstrate a wide range of therapeutic capabilities in the treatment of inflammatory bowel disease (IBD). The intraperitoneal injection of MSCs has exhibited superior therapeutic efficacy on IBD than intravenous injection. Nevertheless, the precise in vivo distribution of MSCs and their biological consequences following intraperitoneal injection remain inadequately understood. Additional studies are required to explore the correlation between MSCs distribution and their biological effects. METHODS First, the distribution of human umbilical cord MSCs (hUC-MSCs) and the numbers of Treg and Th17 cells in mesenteric lymph nodes (MLNs) were analyzed after intraperitoneal injection of hUC-MSCs. Subsequently, the investigation focused on the levels of transforming growth factor beta1 (TGF-β1), a key cytokine to the biology of both Treg and Th17 cells, in tissues of mice with colitis, particularly in MLNs. The study also delved into the impact of hUC-MSCs therapy on Treg cell counts in MLNs, as well as the consequence of TGFB1 knockdown hUC-MSCs on the differentiation of Treg cells and the treatment of IBD. RESULTS The therapeutic effectiveness of intraperitoneally administered hUC-MSCs in the treatment of colitis was found to be significant, which was closely related to their quick migration to MLNs and secretion of TGF-β1. The abundance of hUC-MSCs in MLNs of colitis mice is much higher than that in other organs even the inflamed sites of colon. Intraperitoneal injection of hUC-MSCs led to a significant increase in the number of Treg cells and a decrease in Th17 cells especially in MLNs. Furthermore, the concentration of TGF-β1, the key cytokine for Treg differentiation, were also found to be significantly elevated in MLNs after hUC-MSCs treatment. Knockdown of TGFB1 in hUC-MSCs resulted in a noticeable reduction of Treg cells in MLNs and the eventually failure of hUC-MSCs therapy in colitis. CONCLUSIONS MLNs may be a critical site for the regulatory effect of hUC-MSCs on Treg/Th17 cells and the therapeutic effect on colitis. TGF-β1 derived from hUC-MSCs promotes local Treg differentiation in MLNs. This study will provide new ideas for the development of MSC-based therapeutic strategies in IBD patients.
Collapse
Affiliation(s)
- Qixiang Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhu Zeng
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ning Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, China
| | - Yueyan Su
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, China
| | - Qi Ni
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yukai Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jingwen Yang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaoyan Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huanke Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- , No. 639 Longmian Avenue, Nanjing, Jiangsu, China.
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Tongjiaxiang #24, Nanjing, Jiangsu, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- , No. 639 Longmian Avenue, Nanjing, Jiangsu, China.
| |
Collapse
|
39
|
Zhang Y, Wang C, Li JJ. Revisiting the role of mesenchymal stromal cells in cancer initiation, metastasis and immunosuppression. Exp Hematol Oncol 2024; 13:64. [PMID: 38951845 PMCID: PMC11218091 DOI: 10.1186/s40164-024-00532-4] [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: 01/26/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Immune checkpoint blockade (ICB) necessitates a thorough understanding of intricate cellular interactions within the tumor microenvironment (TME). Mesenchymal stromal cells (MSCs) play a pivotal role in cancer generation, progression, and immunosuppressive tumor microenvironment. Within the TME, MSCs encompass both resident and circulating counterparts that dynamically communicate and actively participate in TME immunosurveillance and response to ICB. This review aims to reevaluate various facets of MSCs, including their potential self-transformation to function as cancer-initiating cells and contributions to the creation of a conducive environment for tumor proliferation and metastasis. Additionally, we explore the immune regulatory functions of tumor-associated MSCs (TA-MSCs) and MSC-derived extracellular vesicles (MSC-EVs) with analysis of potential connections between circulating and tissue-resident MSCs. A comprehensive understanding of the dynamics of MSC-immune cell communication and the heterogeneous cargo of tumor-educated versus naïve MSCs may unveil a new MSC-mediated immunosuppressive pathway that can be targeted to enhance cancer control by ICB.
Collapse
Affiliation(s)
- Yanyan Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Charles Wang
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Jian Jian Li
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA.
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
40
|
Liu D, Liao P, Li H, Tong S, Wang B, Lu Y, Gao Y, Huang Y, Zhou H, Shi L, Papadimitriou J, Zong Y, Yuan J, Chen P, Chen Z, Ding P, Zheng Y, Zhang C, Zheng M, Gao J. Regulation of blood-brain barrier integrity by Dmp1-expressing astrocytes through mitochondrial transfer. SCIENCE ADVANCES 2024; 10:eadk2913. [PMID: 38941455 PMCID: PMC11212732 DOI: 10.1126/sciadv.adk2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/24/2024] [Indexed: 06/30/2024]
Abstract
The blood-brain barrier (BBB) acts as the crucial physical filtration structure in the central nervous system. Here, we investigate the role of a specific subset of astrocytes in the regulation of BBB integrity. We showed that Dmp1-expressing astrocytes transfer mitochondria to endothelial cells via their endfeet for maintaining BBB integrity. Deletion of the Mitofusin 2 (Mfn2) gene in Dmp1-expressing astrocytes inhibited the mitochondrial transfer and caused BBB leakage. In addition, the decrease of MFN2 in astrocytes contributes to the age-associated reduction of mitochondrial transfer efficiency and thus compromises the integrity of BBB. Together, we describe a mechanism in which astrocytes regulate BBB integrity through mitochondrial transfer. Our findings provide innnovative insights into the cellular framework that underpins the progressive breakdown of BBB associated with aging and disease.
Collapse
Affiliation(s)
- Delin Liu
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Sihan Tong
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Bingqi Wang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yafei Lu
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yigang Huang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - Linjing Shi
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China
| | - John Papadimitriou
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Department of Pathology, Pathwest, Nedlands, Western Australia 6009, Australia
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Jun Yuan
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Peilin Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Ziming Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yongqiang Zheng
- Department of Orthopaedics, Jinjiang Municipal Hospital, Jinjiang, Fujian Province, 362200, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Orthopaedics, Jinjiang Municipal Hospital, Jinjiang, Fujian Province, 362200, China
| |
Collapse
|
41
|
Ding P, Gao C, Zhou J, Mei J, Li G, Liu D, Li H, Liao P, Yao M, Wang B, Lu Y, Peng X, Jiang C, Yin J, Huang Y, Zheng M, Gao Y, Zhang C, Gao J. Mitochondria from osteolineage cells regulate myeloid cell-mediated bone resorption. Nat Commun 2024; 15:5094. [PMID: 38877020 PMCID: PMC11178781 DOI: 10.1038/s41467-024-49159-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 05/24/2024] [Indexed: 06/16/2024] Open
Abstract
Interactions between osteolineage cells and myeloid cells play important roles in maintaining skeletal homeostasis. Herein, we find that osteolineage cells transfer mitochondria to myeloid cells. Impairment of the transfer of mitochondria by deleting MIRO1 in osteolineage cells leads to increased myeloid cell commitment toward osteoclastic lineage cells and promotes bone resorption. In detail, impaired mitochondrial transfer from osteolineage cells alters glutathione metabolism and protects osteoclastic lineage cells from ferroptosis, thus promoting osteoclast activities. Furthermore, mitochondrial transfer from osteolineage cells to myeloid cells is involved in the regulation of glucocorticoid-induced osteoporosis, and glutathione depletion alleviates the progression of glucocorticoid-induced osteoporosis. These findings reveal an unappreciated mechanism underlying the interaction between osteolineage cells and myeloid cells to regulate skeletal metabolic homeostasis and provide insights into glucocorticoid-induced osteoporosis progression.
Collapse
Affiliation(s)
- Peng Ding
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Chuan Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Jian Zhou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Jialun Mei
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Gan Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Meng Yao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Bingqi Wang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Yafei Lu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Xiaoyuan Peng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Chenyi Jiang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Jimin Yin
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Yigang Huang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Minghao Zheng
- Centre for Orthopaedic Translational Research, Medical School, University of Western Australia, Nedlands, WA, 6009, Australia
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China.
| |
Collapse
|
42
|
Jiao Q, Xiang L, Chen Y. Mitochondrial transplantation: A promising therapy for mitochondrial disorders. Int J Pharm 2024; 658:124194. [PMID: 38703929 DOI: 10.1016/j.ijpharm.2024.124194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/06/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
As a vital energy source for cellular metabolism and tissue survival, the mitochondrion can undergo morphological or positional change and even shuttle between cells in response to various stimuli and energy demands. Multiple human diseases are originated from mitochondrial dysfunction, but the curative succusses by traditional treatments are limited. Mitochondrial transplantation therapy (MTT) is an innovative therapeutic approach that is to deliver the healthy mitochondria either derived from normal cells or reassembled through synthetic biology into the cells and tissues suffering from mitochondrial damages and finally replace their defective mitochondria and restore their function. MTT has already been under investigation in clinical trials for cardiac ischemia-reperfusion injury and given an encouraging performance in animal models of numerous fatal critical diseases including central nervous system disorders, cardiovascular diseases, inflammatory conditions, cancer, renal injury, and pulmonary damage. This review article summarizes the mechanisms and strategies of mitochondrial transfer and the MTT application for types of mitochondrial diseases, and discusses the potential challenge in MTT clinical application, aiming to exhibit the good therapeutic prospects of MTTs in clinics.
Collapse
Affiliation(s)
- Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Li Xiang
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China; Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China.
| |
Collapse
|
43
|
Chatterjee A, Jayaprakasan M, Chakrabarty AK, Lakkaniga NR, Bhatt BN, Banerjee D, Narwaria A, Katiyar CK, Dubey SK. Comprehensive insights into rheumatoid arthritis: Pathophysiology, current therapies and herbal alternatives for effective disease management. Phytother Res 2024; 38:2764-2799. [PMID: 38522945 DOI: 10.1002/ptr.8187] [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: 11/01/2023] [Revised: 02/14/2024] [Accepted: 03/01/2024] [Indexed: 03/26/2024]
Abstract
Rheumatoid arthritis is a chronic autoimmune inflammatory disease characterized by immune response overexpression, causing pain and swelling in the synovial joints. This condition is caused by auto-reactive antibodies that attack self-antigens due to their incapacity to distinguish between self and foreign molecules. Dysregulated activity within numerous signalling and immunological pathways supports the disease's development and progression, elevating its complexity. While current treatments provide some alleviation, their effectiveness is accompanied by a variety of adverse effects that are inherent in conventional medications. As a result, there is a deep-rooted necessity to investigate alternate therapeutic strategies capable of neutralizing these disadvantages. Medicinal herbs display a variety of potent bioactive phytochemicals that are effective in the complementary management of disease, thus generating an enormous potency for the researchers to delve deep into the development of novel phytomedicine against autoimmune diseases, although additional evidence and understanding are required in terms of their efficacy and pharmacodynamic mechanisms. This literature-based review highlights the dysregulation of immune tolerance in rheumatoid arthritis, analyses the pathophysiology, elucidates relevant signalling pathways involved, evaluates present and future therapy options and underscores the therapeutic attributes of a diverse array of medicinal herbs in addressing this severe disease.
Collapse
Affiliation(s)
- Amrita Chatterjee
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | - Monisha Jayaprakasan
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | | | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
| | | | | | | | | | | |
Collapse
|
44
|
Shan Y, Xie T, Sun Y, Lu Z, Topatana W, Juengpanich S, Chen T, Han Y, Cao J, Hu J, Li S, Cai X, Chen M. Lipid metabolism in tumor-infiltrating regulatory T cells: perspective to precision immunotherapy. Biomark Res 2024; 12:41. [PMID: 38644503 PMCID: PMC11034130 DOI: 10.1186/s40364-024-00588-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/04/2024] [Indexed: 04/23/2024] Open
Abstract
Regulatory T cells (Tregs) are essential to the negative regulation of the immune system, as they avoid excessive inflammation and mediate tumor development. The abundance of Tregs in tumor tissues suggests that Tregs may be eliminated or functionally inhibited to stimulate antitumor immunity. However, immunotherapy targeting Tregs has been severely hampered by autoimmune diseases due to the systemic elimination of Tregs. Recently, emerging studies have shown that metabolic regulation can specifically target tumor-infiltrating immune cells, and lipid accumulation in TME is associated with immunosuppression. Nevertheless, how Tregs actively regulate metabolic reprogramming to outcompete effector T cells (Teffs), and how lipid metabolic reprogramming contributes to the immunomodulatory capacity of Tregs have not been fully discussed. This review will discuss the physiological processes by which lipid accumulation confers a metabolic advantage to tumor-infiltrating Tregs (TI-Tregs) and amplifies their immunosuppressive functions. Furthermore, we will provide a summary of the driving effects of various metabolic regulators on the metabolic reprogramming of Tregs. Finally, we propose that targeting the lipid metabolism of TI-Tregs could be efficacious either alone or in conjunction with immune checkpoint therapy.
Collapse
Affiliation(s)
- Yukai Shan
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Tianao Xie
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Yuchao Sun
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Ziyi Lu
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Win Topatana
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
- School of Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Sarun Juengpanich
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Tianen Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Yina Han
- Department of Pathology, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China
| | - Jiasheng Cao
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Jiahao Hu
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China
| | - Shijie Li
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China.
- School of Medicine, Zhejiang University, 310058, Hangzhou, China.
| | - Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Key Laboratory of Endoscopic Technique Research of Zhejiang Province, No.3 East Qingchun Road, 310016, Hangzhou, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run-Run Shaw Hospital, Zhejiang University, 310016, Hangzhou, China.
- School of Medicine, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
45
|
Jang JH, Zhou M, Makita K, Sun R, El-Hajjar M, Fonseca G, Lauzon AM, Martin JG. Induction of a memory-like CD4 + T-cell phenotype by airway smooth muscle cells. Eur J Immunol 2024; 54:e2249800. [PMID: 38334162 DOI: 10.1002/eji.202249800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
In asthma, CD4+ T-cell interaction with airway smooth muscle (ASM) may enhance its contractile properties and promote its proliferation. However, less is known about the effects of this interaction on T cells. To explore the consequences of interaction of CD4+ T cells with ASM we placed the cells in co-culture and analyzed the phenotypic and functional changes in the T cells. Effector status as well as cytokine expression was assessed by flow cytometry. An increase in CD45RA-CD45RO+ memory T cells was observed after co-culture; however, these cells were not more responsive to CD3/28 restimulation. A reduction in mitochondrial coupling and an increase in the production of mitochondrial reactive oxygen species by CD4+ T cells post-restimulation suggested altered mitochondrial metabolism after co-culture. RNA sequencing analysis of the T cells revealed characteristic downregulation of effector T-cell-associated genes, but a lack of upregulation of memory T-cell-associated genes. The results of this study demonstrate that ASM cells can induce a phenotypic shift in CD4+ T cells into memory-like T cells but with reduced capacity for activation.
Collapse
Affiliation(s)
- Joyce H Jang
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Michael Zhou
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Kosuke Makita
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Mikal El-Hajjar
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Gregory Fonseca
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| | - James G Martin
- Meakins-Christie Laboratories, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
46
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
47
|
Xia Y, Gao D, Wang X, Liu B, Shan X, Sun Y, Ma D. Role of Treg cell subsets in cardiovascular disease pathogenesis and potential therapeutic targets. Front Immunol 2024; 15:1331609. [PMID: 38558816 PMCID: PMC10978666 DOI: 10.3389/fimmu.2024.1331609] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
In the genesis and progression of cardiovascular diseases involving both innate and adaptive immune responses, inflammation plays a pivotal and dual role. Studies in experimental animals indicate that certain immune responses are protective, while others exacerbate the disease. T-helper (Th) 1 cell immune responses are recognized as key drivers of inflammatory progression in cardiovascular diseases. Consequently, the CD4+CD25+FOXP3+ regulatory T cells (Tregs) are gaining increasing attention for their roles in inflammation and immune regulation. Given the critical role of Tregs in maintaining immune-inflammatory balance and homeostasis, abnormalities in their generation or function might lead to aberrant immune responses, thereby initiating pathological changes. Numerous preclinical studies and clinical trials have unveiled the central role of Tregs in cardiovascular diseases, such as atherosclerosis. Here, we review the roles and mechanisms of Treg subsets in cardiovascular conditions like atherosclerosis, hypertension, myocardial infarction and remodeling, myocarditis, dilated cardiomyopathy, and heart failure. While the precise molecular mechanisms of Tregs in cardiac protection remain elusive, therapeutic strategies targeting Tregs present a promising new direction for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunpeng Sun
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dashi Ma
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
48
|
Headley CA, Gautam S, Olmo‐Fontanez A, Garcia‐Vilanova A, Dwivedi V, Akhter A, Schami A, Chiem K, Ault R, Zhang H, Cai H, Whigham A, Delgado J, Hicks A, Tsao PS, Gelfond J, Martinez‐Sobrido L, Wang Y, Torrelles JB, Turner J. Extracellular Delivery of Functional Mitochondria Rescues the Dysfunction of CD4 + T Cells in Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303664. [PMID: 37990641 PMCID: PMC10837346 DOI: 10.1002/advs.202303664] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/17/2023] [Indexed: 11/23/2023]
Abstract
Mitochondrial dysfunction alters cellular metabolism, increases tissue oxidative stress, and may be principal to the dysregulated signaling and function of CD4+ T lymphocytes in the elderly. In this proof of principle study, it is investigated whether the transfer of functional mitochondria into CD4+ T cells that are isolated from old mice (aged CD4+ T cells), can abrogate aging-associated mitochondrial dysfunction, and improve the aged CD4+ T cell functionality. The results show that the delivery of exogenous mitochondria to aged non-activated CD4+ T cells led to significant mitochondrial proteome alterations highlighted by improved aerobic metabolism and decreased cellular mitoROS. Additionally, mito-transferred aged CD4+ T cells showed improvements in activation-induced TCR-signaling kinetics displaying markers of activation (CD25), increased IL-2 production, enhanced proliferation ex vivo. Importantly, immune deficient mouse models (RAG-KO) showed that adoptive transfer of mito-transferred naive aged CD4+ T cells, protected recipient mice from influenza A and Mycobacterium tuberculosis infections. These findings support mitochondria as targets of therapeutic intervention in aging.
Collapse
Affiliation(s)
- Colwyn A. Headley
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
- Biomedical Sciences Graduate ProgramThe Ohio State UniversityColumbusOhio43201USA
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCA94305USA
| | - Shalini Gautam
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | | | | | - Varun Dwivedi
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Anwari Akhter
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Alyssa Schami
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Kevin Chiem
- Disease Intervention & Prevention ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Russell Ault
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
- Biomedical Sciences Graduate ProgramThe Ohio State UniversityColumbusOhio43201USA
| | - Hao Zhang
- Department of Molecular Microbiology and ImmunologySouth Texas Center for Emerging Infectious DiseasesThe University of Texas at San AntonioSan AntonioTX78249USA
| | - Hong Cai
- Department of Molecular Microbiology and ImmunologySouth Texas Center for Emerging Infectious DiseasesThe University of Texas at San AntonioSan AntonioTX78249USA
| | - Alison Whigham
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Jennifer Delgado
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Amberlee Hicks
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Philip S. Tsao
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCA94305USA
| | - Jonathan Gelfond
- UT‐Health San AntonioDepartment of Epidemiology & BiostatisticsSan AntonioTexas78229USA
| | - Luis Martinez‐Sobrido
- Disease Intervention & Prevention ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Yufeng Wang
- Department of Molecular Microbiology and ImmunologySouth Texas Center for Emerging Infectious DiseasesThe University of Texas at San AntonioSan AntonioTX78249USA
| | - Jordi B. Torrelles
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Joanne Turner
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| |
Collapse
|
49
|
Zhai J, Chen Z, Chen P, Yang W, Wei H. Adipose Derived Mesenchymal Stem Cells-Derived Mitochondria Transplantation Ameliorated Erectile Dysfunction Induced by Cavernous Nerve Injury. World J Mens Health 2024; 42:188-201. [PMID: 37382278 PMCID: PMC10782119 DOI: 10.5534/wjmh.220233] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/11/2023] [Accepted: 03/01/2023] [Indexed: 06/30/2023] Open
Abstract
PURPOSE Erectile dysfunction (ED) is a common postoperative complication of pelvic surgery for which there is currently no effective treatment. This study investigated the therapeutic effects and potential mechanisms of adipose derived mesenchymal stem cells-derived mitochondria (ADSCs-mito) transplantation in a rat model of bilateral cavernous nerve injury (CNI) ED. MATERIALS AND METHODS We isolated mitochondria from ADSCs and tested their quality. In vivo, twenty male Sprague Dawley rats were randomly divided into four groups: sham operation group and CNI groups that received intracavernous injection of either phosphate buffer solution, ADSCs-mito or ADSCs. Two weeks after therapy, the erectile function of the rats was evaluated and the penile tissues were harvested for histologic analysis and western blotting. In vitro, the apoptosis rate, reactive oxygen species (ROS), mitochondria derived active oxygen (mtROS) and adenosine triphosphate (ATP) levels were detected in corpus cavernosum smooth muscle cells (CCSMCs) after the incubation with ADSCs-mito. In addition, intercellular mitochondrial transfer was visualized by co-culture of ADSCs and CCSMCs. RESULTS The ADSCs, ADSCs-mito and CCSMCs were isolated and identified successfully. ADSCs-mito transplantation notably restored the erectile function and smooth muscle content of CNI ED rats. Moreover, the levels of ROS, mtROS and cleaved-caspase 3 were reduced and the levels of superoxide dismutase and ATP were increased after ADSCs-mito transplantation. In CNI ED rats, the mitochondrial structure of cells in penile tissues was destroyed. ADSCs could transfer its own mitochondria to CCSMCs. Pre-treatment with ADSCs-mito could significantly decrease apoptosis rate, ROS levels and mtROS levels as well as restore the ATP level in CCSMCs. CONCLUSIONS ADSCs-mito transplantation significantly ameliorated ED induced by CNI, with similar potency to ADSCs treatment. The ADSCs-mito might exert their effects via anti-oxidative stress, anti-apoptosis and modulating energy metabolism of CCSMCs. Mitochondrial transplantation should be a promising therapeutic method for treating CNI ED in the future.
Collapse
Affiliation(s)
- Jiancheng Zhai
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zehong Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Peng Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wende Yang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
50
|
Caicedo A, Morales E, Moyano A, Peñaherrera S, Peña-Cisneros J, Benavides-Almeida A, Pérez-Meza ÁA, Haro-Vinueza A, Ruiz C, Robayo P, Tenesaca D, Barba D, Zambrano K, Castañeda V, Singh KK. Powering prescription: Mitochondria as "Living Drugs" - Definition, clinical applications, and industry advancements. Pharmacol Res 2024; 199:107018. [PMID: 38013162 DOI: 10.1016/j.phrs.2023.107018] [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: 07/09/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
Mitochondria's role as engines and beacons of metabolism and determinants of cellular health is being redefined through their therapeutic application as "Living Drugs" (LDs). Artificial mitochondrial transfer/transplant (AMT/T), encompassing various techniques to modify, enrich, or restore mitochondria in cells and tissues, is revolutionizing acellular therapies and the future of medicine. This article proposes a necessary definition for LDs within the Advanced Therapeutic Medicinal Products (ATMPs) framework. While recognizing different types of LDs as ATMPs, such as mesenchymal stem cells (MSCs) and chimeric antigen receptor T (CAR T) cells, we focus on mitochondria due to their unique attributes that distinguish them from traditional cell therapies. These attributes include their inherent living nature, diverse sources, industry applicability, validation, customizability for therapeutic needs, and their capability to adapt and respond within recipient cells. We trace the journey from initial breakthroughs in AMT/T to the current state-of-the-art applications by emerging innovative companies, highlighting the need for manufacturing standards to navigate the transition of mitochondrial therapies from concept to clinical practice. By providing a comprehensive overview of the scientific, clinical, and commercial landscape of mitochondria as LDs, this article contributes to the essential dialogue among regulatory agencies, academia, and industry to shape their future in medicine.
Collapse
Affiliation(s)
- Andrés Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Sistemas Médicos SIME, Universidad San Francisco de Quito, Quito, Ecuador.
| | - Emilia Morales
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Biología, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Aldana Moyano
- Mito-Act Research Consortium, Quito, Ecuador; Instituto de investigaciones biotecnológicas IIB, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Sebastian Peñaherrera
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Biología, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - José Peña-Cisneros
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Abigail Benavides-Almeida
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Álvaro A Pérez-Meza
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Alissen Haro-Vinueza
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Biología, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Cristina Ruiz
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | | | - Doménica Tenesaca
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Diego Barba
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador
| | - Kevin Zambrano
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Quito, Ecuador; Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands; Instituto de Neurociencias, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Verónica Castañeda
- Universidad San Francisco de Quito USFQ, Instituto de Investigaciones en Biomedicina iBioMed, Quito, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Biología, Colegio de Ciencias Biológicas y Ambientales COCIBA, Universidad San Francisco de Quito USFQ, Quito, Ecuador; PhD Program in Biomedicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Keshav K Singh
- Departments of Genetics, Dermatology and Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|