1
|
Song H, Du X, Zhang Y, Liu W, Luo Y, Liu Y, Xue Y, Xu M, Lu J, Jia W, Du Y, Cao L, Lu J, Zhang W, He Z. Gelatin sponge patch grafting of microcryogel-based three-dimensional mesenchymal stem cells to alleviate acute liver failure. Biomaterials 2025; 321:123324. [PMID: 40253733 DOI: 10.1016/j.biomaterials.2025.123324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/24/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
The clinical application of human umbilical cord mesenchymal stem cells (hUCMSCs) in the treatment of liver failure faces challenges due to cell quality, short engraftment time, and limited efficacy. Here, gelatin microcryogel (GM) microcarriers with pore sizes ranging from 15 to 36 μm were tuned from mixed gelatin and glutaraldehyde to develop a 3D culture system of hUCMSCs with improved therapeutic effects. Bulk RNA sequencing and in vitro assays showed that 3D-hUCMSCs exhibited significant improvement in signaling pathways related to paracrine secretion and anti-inflammation. These 3D-hUCMSCs superior compared to 2D-hUCMSCs not only in terms of paracrine secretion, protection from oxidation, and resistance to mechanical force damage, but also had better liver function improvement effect than 2D-hUCMSCs when they were transplanted as single cells into liver injury mice. Furthermore, a gelatin sponge patch grafting (GSPG) strategy was developed to enable the direct engraftment of 3D-hUCMSCs within the GM microcarriers. The results showed that overall engraftment in the host liver was significantly improved, and the life span of transplanted hosts was extended. Our study provided a practical strategy to achieve high engraftment and long retraining time of 3D-hUCMSCs in rescuing acute liver failure with gelatin matrixes.
Collapse
Affiliation(s)
- Haimeng Song
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Xinyue Du
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Yuanyuan Zhang
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing, 100195, PR China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing, 100195, PR China
| | - Yi Luo
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Yuxin Liu
- Jinzhou Medical University, Jinzhou, Liaoning, 121001, PR China
| | - Yongjia Xue
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Mingyang Xu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China
| | - Jizhen Lu
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, PR China
| | - Yanan Du
- School of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 201619, PR China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 201619, PR China.
| | - Wencheng Zhang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China.
| | - Zhiying He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200123, PR China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, PR China.
| |
Collapse
|
2
|
Williams ME, Banche Niclot F, Rota S, Lim J, Machado J, de Azevedo R, Castillo K, Adebiyi S, Sreenivasan R, Kota D, Mcculloch PC, Taraballi F. Optimizing mesenchymal stem cell therapy: from isolation to GMP-compliant expansion for clinical application. BMC Mol Cell Biol 2025; 26:15. [PMID: 40329219 PMCID: PMC12054297 DOI: 10.1186/s12860-025-00539-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are promising for cell-based therapies targeting a wide range of diseases. However, challenges in translating MSC-based therapies to clinical applications necessitate standardized protocols following Good Manufacturing Practices (GMP) guidelines. This study aimed at developing GMP-complained protocols for FPMSCs isolation and manipulation, necessary for translational research, by (1) optimize culture of MSCs derived from an infrapatellar fat pad (FPMSC) condition through animal-free media comparison and (2) establish feasibility of MSC isolation, manufacturing and storage under GMP-compliance (GMP-FPMSC). METHODS FPMSCs from three different patients were isolated following established protocols and the efficacy of two animal component-free media formulations in the culturing media were evaluated. The impact of different media formulations on cell proliferation, purity, and potency of MSCs was evaluated through doubling time, colony forming unit assay, and percentage of MSCs, respectively. Furthermore, the isolation and expansion of GMP-FPMSCs from four additional donors were optimized and characterized at each stage according to GMP requirements. Viability and sterility were checked using Trypan Blue and Bact/Alert, respectively, while purity and identity were confirmed using Endotoxin, Mycoplasma assays, and Flow Cytometry. The study also included stability assessments post-thaw and viability assessment to determine the shelf-life of the final GMP-FPMSC product. Statistical analyses were conducted using one-way ANOVA with Tukey's Multiple Comparisons. RESULTS The study demonstrated that FPMSCs exhibited enhanced proliferation rates when cultured in MSC-Brew GMP Medium compared to standard MSC media. Cells cultured in this media showed lower doubling times across passages, indicating increased proliferation. Additionally, higher colony formation in FPMSCs cultured in MSC-Brew GMP Medium were observed, supporting enhanced potency. Data from our GMP validation, including cells from 4 different donors, showed post-thaw GMP-FPMSC maintained stem cell marker expression and all the specifications required for product release, including > 95% viability (> 70% is required) and sterility, even after extended storage (up to 180 days), demonstrating the reproducibility and potential of GMP-FPMSCs for clinical use as well as the robustness of the isolation and storage protocols. CONCLUSIONS The study underscores the feasibility of FPMSCs for clinical uses under GMP conditions and emphasizes the importance of optimized culture protocols to improve cell proliferation and potency in MSC-based therapies.
Collapse
Affiliation(s)
- Michael E Williams
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Federica Banche Niclot
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Sara Rota
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jaesang Lim
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Janaina Machado
- Ann Kimball and John W. Johnson Center for Cellular Therapeutics, Houston Methodist Academic Institute, Houston, TX, USA
| | - Ricardo de Azevedo
- Ann Kimball and John W. Johnson Center for Cellular Therapeutics, Houston Methodist Academic Institute, Houston, TX, USA
| | - Katia Castillo
- Ann Kimball and John W. Johnson Center for Cellular Therapeutics, Houston Methodist Academic Institute, Houston, TX, USA
| | - Samuel Adebiyi
- Ann Kimball and John W. Johnson Center for Cellular Therapeutics, Houston Methodist Academic Institute, Houston, TX, USA
| | - Ranga Sreenivasan
- Ann Kimball and John W. Johnson Center for Cellular Therapeutics, Houston Methodist Academic Institute, Houston, TX, USA
| | - Daniel Kota
- Ann Kimball and John W. Johnson Center for Cellular Therapeutics, Houston Methodist Academic Institute, Houston, TX, USA
| | - Patrick C Mcculloch
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA.
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
3
|
Hoang VT, Nguyen QT, Phan TTK, Pham TH, Dinh NTH, Anh LPH, Dao LTM, Bui VD, Dao H, Le DS, Ngo ATL, Le Q, Nguyen Thanh L. Tissue Engineering and Regenerative Medicine: Perspectives and Challenges. MedComm (Beijing) 2025; 6:e70192. [PMID: 40290901 PMCID: PMC12022429 DOI: 10.1002/mco2.70192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/30/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
From the pioneering days of cell therapy to the achievement of bioprinting organs, tissue engineering, and regenerative medicine have seen tremendous technological advancements, offering solutions for restoring damaged tissues and organs. However, only a few products and technologies have received United States Food and Drug Administration approval. This review highlights significant progress in cell therapy, extracellular vesicle-based therapy, and tissue engineering. Hematopoietic stem cell transplantation is a powerful tool for treating many diseases, especially hematological malignancies. Mesenchymal stem cells have been extensively studied. The discovery of induced pluripotent stem cells has revolutionized disease modeling and regenerative applications, paving the way for personalized medicine. Gene therapy represents an innovative approach to the treatment of genetic disorders. Additionally, extracellular vesicle-based therapies have emerged as rising stars, offering promising solutions in diagnostics, cell-free therapeutics, drug delivery, and targeted therapy. Advances in tissue engineering enable complex tissue constructs, further transforming the field. Despite these advancements, many technical, ethical, and regulatory challenges remain. This review addresses the current bottlenecks, emphasizing novel technologies and interdisciplinary research to overcome these hurdles. Standardizing practices and conducting clinical trials will balance innovation and regulation, improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Van T. Hoang
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Quyen Thi Nguyen
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Trang Thi Kieu Phan
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Trang H. Pham
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Nhung Thi Hong Dinh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Le Phuong Hoang Anh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Lan Thi Mai Dao
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Van Dat Bui
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- School of Chemical EngineeringCollege of EngineeringSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Hong‐Nhung Dao
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Duc Son Le
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Anh Thi Lan Ngo
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Quang‐Duong Le
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| |
Collapse
|
4
|
Wang J, Gu Y, Wang J, Zhang N, Han X, Bai Y. Exosomes Derived From BMMSCs Promote B10 Cell Differentiation but Not IL-10 Production. Cell Biochem Funct 2025; 43:e70083. [PMID: 40384415 DOI: 10.1002/cbf.70083] [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: 11/04/2024] [Revised: 04/09/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
The role of IL-10-producing regulatory B cells in inflammatory diseases has recently gained substantial attention. Here, we evaluated the function of mouse bone marrow mesenchymal stem cell-derived exosomes (BMMSC-Exos) and their effect on B-cell differentiation. This study aimed to establish an association between BMMSC-Exos and purified B cells and further explored the anti-inflammatory effect of B10 cells. The expression of inflammatory factors, such as IL-1β, TNF-α, and bone metabolism-related factors, including RANKL, OPG, and secreted IL-10, was investigated by RT-qPCR and ELISA. Populations of CD1dhighCD5+ B cells were analyzed by flow cytometry and immunofluorescence. Cell viability was assessed by CCK8 assay. The results showed that when B cells were separated from BMMSCs by Transwell, IL-1β, TNF-α, and RANKL were downregulated, whereas IL-10, OPG/RANKL, and CD1dhighCD5+ Breg proportion were upregulated in the cocultured groups. Conversely, when B cells were cultured with BMMSC-Exos, increasing concentrations of exosomes increased the proportion of CD1dhighCD5+ and IL-10+CD45+ Bregs; however, the secretion of both pro-inflammatory cytokines and IL-10 were decreased. We found that BMMSC-Exos induce the differentiation of B cells toward the CD1dhighCD5+ and IL-10+CD45+ Breg phenotype but cannot increase the secretion of IL-10 in vitro.
Collapse
Affiliation(s)
- Jiran Wang
- Department of Orthodontics, Beijing Stomatology Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yingzhi Gu
- Department of Orthodontics, Beijing Stomatology Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Jing Wang
- Department of Orthodontics, Beijing Stomatology Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Orthodontics, Beijing Stomatology Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Yuxing Bai
- Department of Orthodontics, Beijing Stomatology Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2025; 70:103-124. [PMID: 38729561 PMCID: PMC11976416 DOI: 10.1016/j.jare.2024.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
6
|
Yan W, Zhang H, Zhang J, Zhao Y, Wu Y, Ma X, Luan X. Human placental mesenchymal stem cells regulate the antioxidant capacity of CD8 +PD-1 + T cells through the CD73/ADO/Nrf2 pathway to protect against liver damage in mice with acute graft-versus-host disease. Mol Immunol 2025; 179:71-83. [PMID: 39923662 DOI: 10.1016/j.molimm.2025.01.016] [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: 12/01/2024] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
Graft-versus-host disease (GVHD) constitutes a severe complication that occurs after allogeneic hematopoietic stem cell transplantation (allo-HSCT), significantly reducing the survival rate of patients. Mesenchymal stem cells (MSCs) are capable of ameliorating the tissue damage caused by GVHD through exerting immunosuppressive effects; however, the relevant mechanisms require further investigation. This study used a GVHD mouse model to explore the therapeutic effects and mechanisms of human placental mesenchymal stem cells (hPMSCs) in mitigating GVHD-induced liver injury. The findings indicated that hPMSCs reduced the proportion of CD8+PD-1+ T cells in both the liver and spleen of GVHD mice, decreased reactive oxygen species (ROS) levels, and upregulated glutathione S transferase (GST) and glutathione (GSH) levels. Consistently, this led to a decrease in the expression of liver fibrosis markers, including alpha-smooth muscle actin (α-SMA) and fibronectin (FN). Moreover, CD8+PD-1+ T cells and ROS were positively correlated with α-SMA and FN, respectively, whereas GST and GSH were negatively correlated with them. hPMSCs with low expression in CD73 attenuated this effect. In vitro studies demonstrated that hPMSCs upregulated the expression of nuclear factor-E2-related factor 2 (Nrf2) via the CD73/adenosine (ADO) pathway, regulated oxidative metabolism, and reduced the number of CD8+PD-1+ T cells. The results suggested that hPMSCs contributed to the regulation of redox homeostasis and reduced the proportion of CD8+PD-1+ T cells through the CD73/ADO/Nrf2 signaling pathway, thereby alleviating liver injury associated with GVHD.
Collapse
Affiliation(s)
- Wei Yan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264000, China; Department of Reproductive Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong Province 264000, China
| | - Hengchao Zhang
- Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Jiashen Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Yaxuan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Yunhua Wu
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Xiaolin Ma
- Hematology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province 266000, China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264000, China.
| |
Collapse
|
7
|
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
|
8
|
Kavaldzhieva K, Mladenov N, Markova M, Belemezova K. Mesenchymal Stem Cell Secretome: Potential Applications in Human Infertility Caused by Hormonal Imbalance, External Damage, or Immune Factors. Biomedicines 2025; 13:586. [PMID: 40149563 PMCID: PMC11940137 DOI: 10.3390/biomedicines13030586] [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: 01/28/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are a source of a wide range of soluble factors, including different proteins, growth factors, cytokines, chemokines, and DNA and RNA molecules, in addition to numerous secondary metabolites and byproducts of their metabolism. MSC secretome can be formally divided into secretory and vesicular parts, both of which are very important for intercellular communication and are involved in processes such as angiogenesis, proliferation, and immunomodulation. Exosomes are thought to have the same content and function as the MSCs from which they are derived, but they also have a number of advantages over stem cells, including low immunogenicity, unaltered functional activity during freezing and thawing, and a lack of tumor formation. In addition, MSC pre-treatment with various inflammatory factors or hypoxia can alter their secretomes so that it can be modified into a more effective treatment. Paracrine factors secreted by MSCs improve the survival of other cell populations by several mechanisms, including immunomodulatory (mostly anti-inflammatory) activity and anti-apoptotic activity partly based on Hsp27 upregulation. Reproductive medicine is one of the fields in which this cell-free approach has been extensively researched. This review presents the possible applications and challenges of using MSC secretome in the treatment of infertility. MSCs and their secretions have been shown to have beneficial effects in various models of female and male infertility resulting from toxic damage, endocrine disorders, trauma, infectious agents, and autoimmune origin.
Collapse
Affiliation(s)
| | | | | | - Kalina Belemezova
- Department of Biology, Medical Faculty, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.K.); (N.M.); (M.M.)
| |
Collapse
|
9
|
Aghajani S, Maboudi SA, Seyhoun I, Nia RR, Shabestari AN, Sharif S, Daneshi M, Verdi J. Review of mesenchymal stem cell-derived exosomes and their potential therapeutic roles in treating rheumatoid arthritis. Mol Biol Rep 2025; 52:229. [PMID: 39948229 DOI: 10.1007/s11033-025-10290-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/22/2025] [Indexed: 05/09/2025]
Abstract
Mesenchymal stem cells (MSCs), one of the most significant categories of stem cells, have garnered considerable attention for their potential in disease treatment due to their unique regenerative properties. MSCs can modulate immune responses through various mechanisms, including the secretion of anti-inflammatory cytokines like IL-10, TGF-β, and extracellular vesicles such as exosomes. The immunomodulatory properties of exosomes have led to their use in treating multiple autoimmune diseases, including rheumatoid arthritis (RA), a common inflammatory joint disease worldwide. Patients with RA experience chronic joint pain, movement disorders, joint and cartilage deformities, and significant treatment costs. The primary treatments for RA consist of pharmacological, non-pharmacological, and surgical methods, which mainly focus on alleviating symptoms and relieving pain rather than offering a complete cure for the disease. Recent clinical trials suggest that cell therapy along with exosome therapy, may be a promising and effective treatment option. Exosomes possess unique features that enable them to transport a variety of medicinal and biological compounds, as well as secrete anti-inflammatory substances and growth factors. Thus, exosomes can help reduce inflammation and pain in patients with rheumatoid arthritis while promoting joint repair and regeneration. In this review, we discuss the remarkable therapeutic effects of MSC-derived exosomes in reducing inflammation, facilitating joint repair, and providing pain relief in RA patients. We also detail the characteristics of MSC-derived exosomes, their isolation techniques, and the pathways of their secretion.
Collapse
Affiliation(s)
- S Aghajani
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - S A Maboudi
- Department of Nano Technology, Tarbiat Modares University, Tehran, Iran
| | - I Seyhoun
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - R Rahim Nia
- Department of NanoMedicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - A Namazi Shabestari
- Department of Geriatric Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sh Sharif
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - M Daneshi
- Department of Medical Laboratory Science, Islamic Azad University Arak Branch, Arak, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Zhang X, Guo Y, Fang K, Huang X, Lan D, Wang M, Jia L, Ji X, Meng R, Zhou D. Therapeutic potential of mesenchymal stem cell-derived extracellular vesicles in ischemic stroke: A meta-analysis of preclinical studies. Brain Res Bull 2025; 221:111219. [PMID: 39837375 DOI: 10.1016/j.brainresbull.2025.111219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/22/2024] [Accepted: 01/17/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Ischemic stroke (IS) remains a significant global health burden, necessitating the development of novel therapeutic strategies. This study aims to systematically evaluate the therapeutic effects of mesenchymal stem cell-derived exosomes (MSC-Exos) on IS outcomes in rodent models. METHODS A comprehensive literature search was conducted across multiple databases to identify studies investigating the effects of MSC-Exos on rodent models of IS. Following rigorous inclusion and exclusion criteria, 73 high-quality studies were selected for meta-analysis. Primary outcomes included reductions in infarct volume/ratio and improvements in functional recovery scores. Data extraction and analysis were performed using RevMan 5.3 software. RESULTS Pooled data indicated that MSC-Exos administration significantly reduced infarct size and improved functional recovery scores in rodent models of IS. Treatment within 24 hours and beyond 24 hours of stroke induction both demonstrated substantial reductions in infarct volume/ratio compared to controls. Furthermore, MSC-Exos-treated groups exhibited marked improvements in functional recovery, as assessed by various neurobehavioral tests. The meta-analysis showed no significant publication bias, and heterogeneity levels were acceptable. CONCLUSIONS MSC-Exos reveal significant therapeutic potential for IS, with evidence supporting their efficacy in reducing infarct size and enhancing functional recovery in preclinical rodent models. These findings pave the way for further research and potential clinical translation.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Yibing Guo
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Kun Fang
- Capital Medical University, Beijing 100069, China.
| | - Xiangqian Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Duo Lan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Mengqi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Lina Jia
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
11
|
Ji W, Xu J, Huang C, Liu T, Chen S, Zhao Y, Zhou C, Sun L, Wang M, Wang D, Zhu W. Gastric cancer mesenchymal stem cells upregulate PD-1 expression on the CD8 + T cells by regulating the PI3K/AKT pathway. Mol Immunol 2025; 178:97-106. [PMID: 39870015 DOI: 10.1016/j.molimm.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/27/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
Gastric cancer mesenchymal stem cells (GC-MSCs) are a crucial component of the gastric cancer microenvironment, exerting a pivotal influence on the formation of a suppressive immune microenvironment and the progression of gastric cancer. In this study, we utilized GC-MSCs to co-culture peripheral blood mononuclear cells (PBMCs) obtained from both gastric cancer patients and healthy individuals in a proportionate manner by direct cell-to-cell contact. Our findings reveal that co-culture of GC-MSCs with PBMCs led to a notable reduction in CD8+ T cells percentages and an increase in surface PD-1 expression levels on CD8+ T cells. However, flow cytometry analysis demonstrated no significant changes following pretreatment with GC-MSC-conditioned medium (GC-MSC-CM). Moreover, western blotting analysis demonstrated a notable reduction in phosphorylated AKT levels in co-cultured PBMCs. Collectively, our results suggest that GC-MSCs impair the anti-tumor immune response of PBMCs by elevating the PD-1 levels of CD8+ T cells and impairing the killing of CD8+ T cells. These findings provide new evidence that GC-MSCs contribute to immunosuppression within the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Weimeng Ji
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu 212001, China; Department of Oncology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Juan Xu
- Department of Laboratory Medicine, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Chao Huang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Ting Liu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Shihan Chen
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Chenglin Zhou
- Department of Laboratory Medicine, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Li Sun
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of jiangsu University, Suzhou, Jiangsu 215399, China
| | - Mei Wang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Deqiang Wang
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu 212001, China; Department of Oncology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China.
| | - Wei Zhu
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu 212001, China; Department of Oncology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
12
|
Cohen DJ, Van Duyn CM, Deng J, Lodi MK, Gallagher MB, Sugar JT, Rawlinson JJ, Ghosh P, Boyan BD, Schwartz Z. Bone Marrow Stromal Cells Generate a Pro-Healing Inflammasome When Cultured on Titanium-Aluminum-Vanadium Surfaces with Microscale/Nanoscale Structural Features. Biomimetics (Basel) 2025; 10:66. [PMID: 39851782 PMCID: PMC11759188 DOI: 10.3390/biomimetics10010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The surface topography and chemistry of titanium-aluminum-vanadium (Ti6Al4V) implants play critical roles in the osteoblast differentiation of human bone marrow stromal cells (MSCs) and the creation of an osteogenic microenvironment. To assess the effects of a microscale/nanoscale (MN) topography, this study compared the effects of MN-modified, anodized, and smooth Ti6Al4V surfaces on MSC response, and for the first time, directly contrasted MN-induced osteoblast differentiation with culture on tissue culture polystyrene (TCPS) in osteogenic medium (OM). Surface characterization revealed distinct differences in microroughness, composition, and topography among the Ti6Al4V substrates. MSCs on MN surfaces exhibited enhanced osteoblastic differentiation, evidenced by increased expression of RUNX2, SP7, BGLAP, BMP2, and BMPR1A (fold increases: 3.2, 1.8, 1.4, 1.3, and 1.2). The MN surface also induced a pro-healing inflammasome with upregulation of anti-inflammatory mediators (170-200% increase) and downregulation of pro-inflammatory factors (40-82% reduction). Integrin expression shifted towards osteoblast-associated integrins on MN surfaces. RNA-seq analysis revealed distinct gene expression profiles between MSCs on MN surfaces and those in OM, with only 199 shared genes out of over 1000 differentially expressed genes. Pathway analysis showed that MN surfaces promoted bone formation, maturation, and remodeling through non-canonical Wnt signaling, while OM stimulated endochondral bone development and mineralization via canonical Wnt3a signaling. These findings highlight the importance of Ti6Al4V surface properties in directing MSC differentiation and indicate that MN-modified surfaces act via signaling pathways that differ from OM culture methods, more accurately mimicking peri-implant osteogenesis in vivo.
Collapse
Affiliation(s)
- David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Christine M. Van Duyn
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Jingyao Deng
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
| | - Musaddiq K. Lodi
- Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | | | - James T. Sugar
- Medtronic Spine, Memphis, TN 38132, USA; (M.B.G.); (J.T.S.); (J.J.R.)
| | | | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
- Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (D.J.C.); (C.M.V.D.); (J.D.); (Z.S.)
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
13
|
Veret D, Tejedor G, Perez E, Chomette A, Farno M, Ferreira-Lopez R, Dagneaux L, Pers YM, Jorgsensen C, Gondeau C, Brondello JM. Combination of rapamycin and adipose-derived mesenchymal stromal cells enhances therapeutic potential for osteoarthritis. Stem Cell Res Ther 2025; 16:9. [PMID: 39815291 PMCID: PMC11737215 DOI: 10.1186/s13287-024-04090-8] [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] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The regenerative potential of mesenchymal stromal/stem cells (MSCs) has been extensively studied in clinical trials in the past decade. However, despite the promising regenerative properties documented in preclinical studies, for instance in osteoarthritis (OA), the therapeutic translation of these results in patients has not been fully conclusive. One factor contributing to this therapeutic barrier could be the presence of senescent cells in OA joints. METHODS This study evaluated a novel approach to OA treatment by combining adipose tissue-derived MSCs (AD-MSCs) with rapamycin, a clinically approved immunosuppressive drug with anti-senescence properties. First, rapamycin effects on senescence and fibrosis markers were investigated in freshly isolated OA chondrocytes by immunostaining. Next, the in vitro differentiation capacities of AD-MSCs, their regulatory immune functions on activated immune cells and their regenerative effects on OA chondrocyte signature were assessed in the presence of rapamycin. RESULTS In OA chondrocytes, rapamycin reduced the senescence marker p15INK4B and the fibrosis marker COL1A1 without affecting the expression of the master chondrogenic markers SOX9 and COL2. Rapamycin also enhanced AD-MSC differentiation into chondrocytes and reduced their differentiation into adipocytes. In addition, rapamycin improved AD-MSC immunoregulatory functions by promoting the expression of immunosuppressive factors, such as IDO1, PTGS2 and also CD274 (encoding PD-L1). Finally, RNA sequencing analysis showed that in the presence of rapamycin, AD-MSCs displayed improved chondroprotective regenerative effects on co-cultured OA chondrocytes. CONCLUSIONS Our findings suggest that the rapamycin and AD-MSC combination enhances the therapeutic efficacy of these cells in senescence-driven degenerative diseases such as OA, notably by improving their anti-fibrotic and anti-inflammatory properties.
Collapse
Affiliation(s)
- Damien Veret
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | | | - Esther Perez
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | | | - Maylis Farno
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | - Rosanna Ferreira-Lopez
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
- Rheumatology department, Regional Narbonne Hospital, Narbonne, France
| | - Louis Dagneaux
- Hôpital Lapeyronie, Orthopedic Service, Montpellier, France
| | - Yves-Marie Pers
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Christian Jorgsensen
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Claire Gondeau
- MedXCell, IRMB, CHU St Eloi, Cyborg, Montpellier, France
| | - Jean-Marc Brondello
- IRMB, Univ Montpellier, INSERM, CHU St Eloi, 80 AV A Fliche, 34295-Cedex-05, Montpellier, France.
| |
Collapse
|
14
|
Shahrezaei A, Sohani M, Nasirinezhad F. Mesenchymal stem cells as a therapeutic strategy to combat oxidative stress-mediated neuropathic pain. BIOIMPACTS : BI 2025; 15:30648. [PMID: 40256229 PMCID: PMC12008502 DOI: 10.34172/bi.30648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 04/22/2025]
Abstract
Neuropathic pain, a chronic condition resulting from somatosensory system damage, remains a significant clinical challenge due to its complex pathophysiology and inadequate response to traditional therapies. Oxidative stress, characterized by an imbalance between free radicals production and antioxidant defenses, plays a pivotal role in the development and maintenance of neuropathic pain. Mesenchymal stem cells (MSCs) are multipotent stromal cells with the ability to differentiate into various cell types and possess immunomodulatory, anti-inflammatory, and regenerative properties, making them promising candidates for novel pain management strategies. Preclinical studies demonstrate that MSCs can reduce inflammation, scavenge reactive oxygen species (ROS), promote nerve regeneration, and modulate pain signaling pathways. Various administration routes, including intravenous and intrathecal, have been investigated to optimize MSC delivery and efficacy. Additionally, MSC-derived extracellular vesicles (EVs) represent a cell-free alternative with substantial therapeutic potential. Despite encouraging preclinical findings, further research is needed to refine MSC-based therapies, including the exploration of combination treatments and rigorous clinical trials, to translate these promising results into effective clinical applications for neuropathic pain relief. This review explores the therapeutic potential of MSCs in alleviating oxidative stress-mediated neuropathic pain.
Collapse
Affiliation(s)
- Aidin Shahrezaei
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sohani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farinaz Nasirinezhad
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Center of Experimental and Comparative Study, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Zhao DZ, Yang RL, Wei HX, Yang K, Yang YB, Wang NX, Zhang Q, Chen F, Zhang T. Advances in the research of immunomodulatory mechanism of mesenchymal stromal/stem cells on periodontal tissue regeneration. Front Immunol 2025; 15:1449411. [PMID: 39830512 PMCID: PMC11739081 DOI: 10.3389/fimmu.2024.1449411] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Periodontal disease is a highly prevalent disease worldwide that seriously affects people's oral health, including gingivitis and periodontitis. Although the current treatment of periodontal disease can achieve good control of inflammation, it is difficult to regenerate the periodontal supporting tissues to achieve a satisfactory therapeutic effect. In recent years, due to the good tissue regeneration ability, the research on Mesenchymal stromal/stem cells (MSCs) and MSC-derived exosomes has been gradually deepened, especially its ability to interact with the microenvironment of the body in the complex immunoregulatory network, which has led to many new perspectives on the therapeutic strategies for many diseases. This paper systematically reviews the immunomodulatory (including bone immunomodulation) properties of MSCs and their role in the periodontal inflammatory microenvironment, summarizes the pathways and mechanisms by which MSCs and MSC-EVs have promoted periodontal regeneration in recent years, lists potential areas for future research, and describes the issues that should be considered in future basic research and the direction of development of "cell-free therapies" for periodontal regeneration.
Collapse
Affiliation(s)
- De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Rui-Lin Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Han-Xiao Wei
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kang Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yi-Bing Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Nuo-Xin Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Chen
- Department of Prosthetics, Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
16
|
Rasouli M, Alavi M, D'Angelo A, Sobhani N, Roudi R, Safari F. Exploring the dichotomy of the mesenchymal stem cell secretome: Implications for tumor modulation via cell-signaling pathways. Int Immunopharmacol 2024; 143:113265. [PMID: 39353385 DOI: 10.1016/j.intimp.2024.113265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Current cancer therapeutic strategies for the treatment of cancer are often unsuccessful due to unwanted side effects and drug resistance. Therefore, the design and development of potent, new anticancer platforms, such as stem-cell treatments, have attracted much attention. Distinctive biological properties of stem cells include their capacity to secrete bioactive factors, their limited immunogenicity, and their capacity for renewing themselves. Mesenchymal stem cells (MSCs) are one of several kinds of stem cells that are conveniently extracted and are able to be cultivated in vitro utilizing various sources. The secretome of stem cells contains many trophic factors, including cytokines, chemokines, growth factors, and microRNA molecules that can either promote or inhibit the formation of tumors, based on the cell environment. In the current review, we focused on the secretome of mesenchymal stem cells. These stem cells act as a double-edged sword in the regulation of cell signal transduction pathways in that they can either suppress or promote tumors.
Collapse
Affiliation(s)
- Mohammad Rasouli
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mana Alavi
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Alberto D'Angelo
- Oncology Department, Royal United Hospital, Bath BA1 3NG, United Kingdom
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA.
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| |
Collapse
|
17
|
Roseti L, Cavallo C, Desando G, D’Alessandro M, Grigolo B. Forty Years of the Use of Cells for Cartilage Regeneration: The Research Side. Pharmaceutics 2024; 16:1622. [PMID: 39771600 PMCID: PMC11677864 DOI: 10.3390/pharmaceutics16121622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The treatment of articular cartilage damage has always represented a problem of considerable practical interest for orthopedics. Over the years, many surgical techniques have been proposed to induce the growth of repairing tissue and limit degeneration. In 1994, the turning point occurred: implanted autologous cells paved the way for a new treatment option based more on regeneration than repair. Objectives: This review aims to outline biological and clinical advances, from the use of mature adult chondrocytes to cell-derived products, going through progenitor cells derived from bone marrow or adipose tissue and their concentrates for articular cartilage repair. Moreover, it highlights the relevance of gene therapy as a valuable tool for successfully implementing current regenerative treatments, and overcoming the limitations of the local delivery of growth factors. Conclusions: Finally, this review concludes with an outlook on the importance of understanding the role and mechanisms of action of the different cell compounds with a view to implementing personalized treatments.
Collapse
Affiliation(s)
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.); (G.D.); (M.D.); (B.G.)
| | | | | | | |
Collapse
|
18
|
Sharun K, Banu SA, Alifsha B, Abualigah L, Pawde AM, Dhama K, Pal A. Mesenchymal stem cell therapy in veterinary ophthalmology: clinical evidence and prospects. Vet Res Commun 2024; 48:3517-3531. [PMID: 39212813 DOI: 10.1007/s11259-024-10522-w] [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: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mesenchymal stem cell (MSC) therapy presents a promising strategy for treating various ocular conditions in veterinary medicine. This review explores the therapeutic potential of MSCs in managing corneal ulcers, immune-mediated keratitis, chronic superficial keratitis, keratoconjunctivitis sicca, retinal degeneration, and ocular burns in feline, equine, and canine patients. Studies have demonstrated the immunomodulatory and regenerative properties of MSCs, highlighting their ability to mitigate inflammation and promote tissue regeneration. Experimental studies have shown the potential of MSC therapy in reducing corneal opacity and vascularization, indicating significant therapeutic advantages. Delivery methods play a crucial role in optimizing the therapeutic efficacy of MSCs in ocular diseases. Various delivery methods, such as intravitreal injection, subconjunctival injection, topical administration, and scaffold-mediated delivery, are being explored to optimize MSC delivery to the target ocular tissues. Clinical trials have shown significant improvements in clinical signs following MSC therapy, underscoring its efficacy in treating ocular diseases. Additionally, tissue engineering approaches incorporating MSCs, growth factors, and scaffolds offer innovative strategies for corneal regeneration and tissue repair. Despite challenges such as standardization of protocols and long-term safety assessment, ongoing research endeavours seek to unlock the full therapeutic potential of MSC therapy in ocular diseases. Future prospects in MSC therapy involve exploring scaffold and hydrogel-based approaches and cell-free therapies leveraging the bioactive molecules released by MSCs. Continued research and development efforts are essential to unlock the full therapeutic potential of MSCs and realize their transformative impact on ocular diseases in veterinary patients.
Collapse
Affiliation(s)
- Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - S Amitha Banu
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - B Alifsha
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laith Abualigah
- Computer Science Department, Al al-Bayt University, Mafraq, 25113, Jordan
- MEU Research Unit, Middle East University, Amman, 11831, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, 11931, Jordan
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
- Artificial Intelligence and Sensing Technologies (AIST) Research Center, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - A M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Amar Pal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
19
|
Ginting AR, Munir D, Amin MM, Darlan DM, Putra A, Rusda M, Mutiara E, Mayasari E, Rozi MF. Mesenchymal stem cells for immune modulation in systemic lupus erythematosus: From bench research to clinical applications. NARRA J 2024; 4:e994. [PMID: 39816093 PMCID: PMC11731813 DOI: 10.52225/narra.v4i3.994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/15/2024] [Indexed: 01/18/2025]
Abstract
Systemic lupus erythematosus (SLE) is a prevalent autoimmune disease affecting multiple organ systems. Disease progression is inevitable as part of its natural course, necessitating aggressive therapeutic strategies, particularly with the use of immunosuppressants. Long-term use of steroids and other immunosuppressants is associated with significant adverse effects. Mesenchymal stem cells (MSCs) have been shown to modulate the immune response, leading to immunosuppressive effects against self-antigens. MSCs have demonstrated the ability to modulate several immune cell populations, contributing to favorable outcomes in controlling immune and inflammatory conditions. Recent evidence has shown an increase in Treg and Breg cell subsets following MSC administration, along with modulation of other immune cells, including dendritic cells, B cells, and T cells. However, the balance between MSC pro-inflammatory and anti-inflammatory phenotypic activation remains a critical factor in determining therapeutic outcomes. Various covariates also influence the efficacy of MSC therapy. The aim of this study was to provide a comprehensive overview of the utilization of mesenchymal stem cells (MSCs) in SLE treatment, leveraging their immunomodulatory and immunosuppressive capabilities. Understanding the fundamental preclinical effects of MSCs and recent findings from clinical studies may enhance the potential of MSC therapy in the management of SLE patients.
Collapse
Affiliation(s)
- Andi R. Ginting
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Delfitri Munir
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Pusat Unggulan Iptek Tissue Engineering, Universitas Sumatera Utara, Medan, Indonesia
| | - Mustafa M. Amin
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Dewi M. Darlan
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research Center, Semarang, Indonesia
- Faculty of Postgraduate Biomedical Science, Universitas Islam Sultan Agung, Semarang, Indonesia
| | - Muhammad Rusda
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Erna Mutiara
- Faculty of Public Health, Universitas Sumatera Utara, Medan, Indonesia
| | - Evita Mayasari
- Department of Microbiology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad F. Rozi
- Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
20
|
Liu H, Cui D, Huangfu S, Wang X, Yu X, Yang H, Zheng X, Li Y, Bi J, Zhang L, Wang P. VCAM-1 + Mesenchymal Stem/Stromal Cells Reveal Preferable Efficacy Upon an Experimental Autoimmune Encephalomyelitis Mouse Model of Multiple Sclerosis Over the VCAM-1 - Counterpart. Neurochem Res 2024; 50:40. [PMID: 39613932 PMCID: PMC11607028 DOI: 10.1007/s11064-024-04267-w] [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: 04/05/2024] [Revised: 09/07/2024] [Accepted: 09/24/2024] [Indexed: 12/01/2024]
Abstract
Despite the considerable progress in mesenchymal stem/stromal cells (MSCs)-based novel intervention of multiple sclerosis (MS), yet the disease-modifying effect of VCAM-1- MSCs and novel VCAM-1+ counterpart is largely obscure. In this study, we took advantage of the EAE mouse model and VCAM-1+ human umbilical cord-derived MSCs (hUC-MSCs) for the evaluation of the therapeutic effect of systematic MSCs infusion. On the one hand, we compared the protective effect of VCAM-1- and VCAM-1+ hUC-MSCs against the clinical symptoms, demyelination, active glia cells and neuroinflammation in EAE mice by conducting multifaceted detections upon spinal cord and brain tissues. On the other hand, we conducted RNA-sequencing (RNA-SEQ) and multidimensional bioinformatics analyses for the evaluation of the transcriptomic features of spinal cord tissue in EAE mice after systematic hUC-MSCs infusion. Compared to those with VCAM-1- hUC-MSCs injection, VCAM-1+ mice showed further remission in clinical manifestations, and in particular, the inflammatory infiltration and active glial cells. Mice in all groups revealed conservations in overall gene expression profiling and somatic mutation spectrum. The differentially expressed genes (DEGs) between EAE mice and those with hUC-MSCs infusion were mainly involved in neuroinflammation and inflammatory response. Our findings indicated the feasibility of VCAM-1+ hUC-MSCs for multiple sclerosis treatment, which would supply new references for the development of novel VCAM-1+ MSCs-based cytotherapy in future.
Collapse
Affiliation(s)
- Haixia Liu
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
- Department of Neurology, Weihai Municipal Hospital, Weihai, China
| | - Dongqing Cui
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
| | - Shasha Huangfu
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
| | - Xiaojun Wang
- Universal Biomedical Research Institute, Zibo, China
| | - Xiao Yu
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
| | - Hui Yang
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
| | - Xiaolei Zheng
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
| | - Yan Li
- Universal Biomedical Research Institute, Zibo, China
| | - Jianzhong Bi
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China
| | - Leisheng Zhang
- Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Blood Ecology and Biointelligence, Jinan Key Laboratory of Medical Cell Bioengineering, The Fourth People's Hospital of Jinan, The Teaching Hospital of Shandong First Medical University, 50 Shifan Road, Jinan, China.
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, 204 Donggang West Road, Lanzhou, China.
| | - Ping Wang
- Department of Neurology, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, China.
| |
Collapse
|
21
|
Aghayan AH, Mirazimi Y, Nasehi L, Atashi A. The toxic effects of neutrophil extracellular traps on mesenchymal stem cells. Mol Biol Rep 2024; 52:30. [PMID: 39614028 DOI: 10.1007/s11033-024-10134-2] [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: 04/15/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Sepsis, a systemic inflammatory response syndrome resulting from an uncontrolled inflammatory reaction to infection, remains without a definitive cure despite therapeutic advancements. Mesenchymal stem cells (MSCs), renowned for their capacity to alleviate inflammation and modulate the immune system, have emerged as a potential treatment avenue for sepsis. In sepsis pathophysiology, hyperactivated neutrophils release extracellular neutrophil traps (NETs). NETs are essential for eradicating pathogens; however, excessive formation leads to tissue damage. Given the limited knowledge regarding the impact of NETs on MSCs used in sepsis therapy and the established interaction between MSCs and NETs, this study investigates the effects of NETs on MSCs in vitro. NETs were isolated from stimulated neutrophils, and MSCs were sourced from umbilical cord blood. After co-culturing MSCs with isolated NETs, MSCs' viability, migration, intracellular antioxidant capacity, and changes in gene expression were analyzed. Following exposure to NETs, MSCs exhibited obvious apoptosis and necrosis. NETs disrupt MSCs' mitochondrial activity. Also, NETs upregulate the pro-apoptotic gene BAX and downregulate the anti-apoptotic gene BCL2 in MSCs. Additionally, NETs reduce MSCs' intracellular antioxidant capacity. Furthermore, MSC migration is significantly impaired by NETs. This study collectively demonstrates that NETs have toxic and detrimental effects on MSCs. These effects on MSCs indicate a potential barrier to their functionality and therapeutic efficacy. Therefore, it appears that reducing the undesirable effects of NETs could serve as a novel target to enhance the therapeutic efficacy of MSCs in septic patients.
Collapse
Affiliation(s)
- Amir Hossein Aghayan
- Student Research Committee, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Yasin Mirazimi
- Student Research Committee, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Leila Nasehi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Amir Atashi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran.
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
22
|
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
|
23
|
Colonne CK, Kimble EL, Turtle CJ. Evolving strategies to overcome barriers in CAR-T cell therapy for acute myeloid leukemia. Expert Rev Hematol 2024; 17:797-818. [PMID: 39439295 DOI: 10.1080/17474086.2024.2420614] [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: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex and heterogeneous disease characterized by an aggressive clinical course and limited efficacious treatment options in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy is an investigational treatment strategy for R/R AML that has shown some promise. However, obstacles to successful CAR-T cell immunotherapy for AML remain. AREAS COVERED In analyses of clinical trials of CAR-T cell therapy for R/R AML, complete responses without measurable residual disease have been reported, but the durability of those responses remains unclear. Significant barriers to successful CAR-T cell therapy in AML include the scarcity of suitable tumor-target antigens (TTA), inherent T cell functional deficits, and the immunoinhibitory and hostile tumor microenvironment (TME). This review will focus on these barriers to successful CAR-T cell therapy in AML, and discuss scientific advancements and evolving strategies to overcome them. EXPERT OPINION Achieving durable remissions in R/R AML will likely require a multifaceted approach that integrates advancements in TTA selection, enhancement of the intrinsic quality of CAR-T cells, and development of strategies to overcome inhibitory mechanisms in the AML TME.
Collapse
Affiliation(s)
- Chanukya K Colonne
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erik L Kimble
- Translational Science and Therapeutic Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Cameron J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
24
|
Castilla-Casadiego DA, Loh DH, Pineda-Hernandez A, Rosales AM. Stimuli-Responsive Substrates to Control the Immunomodulatory Potential of Stromal Cells. Biomacromolecules 2024; 25:6319-6337. [PMID: 39283807 PMCID: PMC11506505 DOI: 10.1021/acs.biomac.4c00835] [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] [Indexed: 10/06/2024]
Abstract
Mesenchymal stromal cells (MSCs) have broad immunomodulatory properties that range from regulation, proliferation, differentiation, and immune cell activation to secreting bioactive molecules that inhibit inflammation and regulate immune response. These properties provide MSCs with high therapeutic potency that has been shown to be relevant to tissue engineering and regenerative medicine. Hence, researchers have explored diverse strategies to control the immunomodulatory potential of stromal cells using polymeric substrates or scaffolds. These substrates alter the immunomodulatory response of MSCs, especially through biophysical cues such as matrix mechanical properties. To leverage these cell-matrix interactions as a strategy for priming MSCs, emerging studies have explored the use of stimuli-responsive substrates to enhance the therapeutic value of stromal cells. This review highlights how stimuli-responsive materials, including chemo-responsive, microenvironment-responsive, magneto-responsive, mechano-responsive, and photo-responsive substrates, have specifically been used to promote the immunomodulatory potential of stromal cells by controlling their secretory activity.
Collapse
Affiliation(s)
- David A Castilla-Casadiego
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Darren H Loh
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Aldaly Pineda-Hernandez
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Adrianne M Rosales
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
25
|
Desai S, Sharma D, Srinivas R, Balaji V, Thakore V, Bedi VS, Jindal R, Sugumaran A, Mohanasundaram S, Gogtay J, Gupta PK, Bhuiyan A, Atturu G. Mesenchymal stromal cell therapy (REGENACIP ®), a promising treatment option in chronic limb threatening ischemia - a narrative review. Stem Cell Res Ther 2024; 15:352. [PMID: 39380065 PMCID: PMC11463160 DOI: 10.1186/s13287-024-03957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
Chronic Limb Threatening Ischemia (CLTI) is a challenging clinical problem associated with high morbidity and mortality. Endovascular interventions have been the cornerstone of treatment whenever possible. It is estimated that CLTI represents < 10% of all Peripheral Artery Disease patients, yet 50% of the patients end up either with a major amputation of the lower limbs or die of cardiovascular causes within one year period, especially in those with unsuccessful revascularization or "no-option" CLTI. Cell-based therapeutics, especially bone marrow-derived mesenchymal stromal cells have emerged as a potential, promising, and novel alternate therapeutic modality in the management of CLTI, bolstered with positive results in numerous research, including randomized and nonrandomized trials. REGENACIP® is one such BM-MSC therapy approved by Central Drugs Standard Control Organization in India for the management of "no-option" Atherosclerotic Peripheral Arterial disease / Buerger's disease patients with established critical limb ischemia in Rutherford Grade III-5 or III-6, not eligible for or have failed traditional revascularization treatment, with rest pain and / or ulcers in the affected limb. The current review aims to deliberate upon the various aspects of CLTI and clinical benefits of REGENACIP® therein.
Collapse
Affiliation(s)
- Sanjay Desai
- Senior Consultant Vascular and Endovascular surgeon, Ramaiah Memorial Hospital, Bengaluru, India
| | - Digvijay Sharma
- Head of Department, Vascular Interventions and Surgery, Fortis Escorts Heart Institute, New Delhi, India
| | - Rajesh Srinivas
- Vascular Surgeon, NH-Mazumdar-Shaw Medical Center, Bengaluru, India
| | | | - Vijay Thakore
- Senior Vascular Surgeon, Aadicura Super Speciality Hospitals, Vadodara, India
| | - Varinder Singh Bedi
- Chairman & Senior Consultant, Institute of Vascular & Endovascular Sciences, Sir Ganga Ram Hospital, New Delhi, India
| | - Ravul Jindal
- Director of Vascular & Endovascular Surgery, Fortis Hospital, Mohali, India
| | | | | | | | | | - Aniruddha Bhuiyan
- Consultant Vascular & Endovascular Surgery, Vascular Care n Cure, Mumbai, India
| | - Gnaneswar Atturu
- Head & Senior Consultant, Department of Vascular & Endovascular Surgery, Renova Hospitals, Hyderabad, India
| |
Collapse
|
26
|
D’Angeli F, Granata G, Romano IR, Distefano A, Lo Furno D, Spila A, Leo M, Miele C, Ramadan D, Ferroni P, Li Volti G, Accardo P, Geraci C, Guadagni F, Genovese C. Biocompatible Poly(ε-Caprolactone) Nanocapsules Enhance the Bioavailability, Antibacterial, and Immunomodulatory Activities of Curcumin. Int J Mol Sci 2024; 25:10692. [PMID: 39409022 PMCID: PMC11476408 DOI: 10.3390/ijms251910692] [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: 09/08/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Curcumin (Cur), the primary curcuminoid found in Curcuma longa L., has garnered significant attention for its potential anti-inflammatory and antibacterial properties. However, its hydrophobic nature significantly limits its bioavailability. Additionally, adipose-derived stem cells (ADSCs) possess immunomodulatory properties, making them useful for treating inflammatory and autoimmune conditions. This study aims to verify the efficacy of poly(ε-caprolactone) nanocapsules (NCs) in improving Cur's bioavailability, antibacterial, and immunomodulatory activities. The Cur-loaded nanocapsules (Cur-NCs) were characterized for their physicochemical properties (particle size, polydispersity index, Zeta potential, and encapsulation efficiency) and stability over time. A digestion test simulated the behavior of Cur-NCs in the gastrointestinal tract. Micellar phase analyses evaluated the Cur-NCs' bioaccessibility. The antibacterial activity of free Cur, NCs, and Cur-NCs against various Gram-positive and Gram-negative strains was determined using the microdilution method. ADSC viability, treated with Cur-NCs and Cur-NCs in the presence or absence of lipopolysaccharide, was analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide assay. Additionally, ADSC survival was assessed through the Muse apoptotic assay. The expression of both pro-inflammatory (interleukin-1β and tumor necrosis factor-α) and anti-inflammatory (IL-10 and transforming growth factor-β) cytokines on ADSCs was evaluated by real-time polymerase chain reaction. The results demonstrated high stability post-gastric digestion of Cur-NCs and elevated bioaccessibility of Cur post-intestinal digestion. Moreover, Cur-NCs exhibited antibacterial activity against Escherichia coli without affecting Lactobacillus growth. No significant changes in the viability and survival of ADSCs were observed under the experimental conditions. Finally, Cur-NCs modulated the expression of both pro- and anti-inflammatory cytokines in ADSCs exposed to inflammatory stimuli. Collectively, these findings highlight the potential of Cur-NCs to enhance Cur's bioavailability and therapeutic efficacy, particularly in cell-based treatments for inflammatory diseases and intestinal dysbiosis.
Collapse
Affiliation(s)
- Floriana D’Angeli
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Giuseppe Granata
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123 Catania, Italy; (I.R.R.); (D.L.F.)
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy; (A.D.); (G.L.V.)
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123 Catania, Italy; (I.R.R.); (D.L.F.)
| | - Antonella Spila
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Mariantonietta Leo
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Chiara Miele
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Dania Ramadan
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
| | - Patrizia Ferroni
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
- InterInstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele, 00166 Rome, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy; (A.D.); (G.L.V.)
| | - Paolo Accardo
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Corrada Geraci
- CNR-Institute of Biomolecular Chemistry, Via Paolo Gaifami 18, 95126 Catania, Italy; (G.G.); (P.A.); (C.G.)
| | - Fiorella Guadagni
- Department of Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.S.); (M.L.); (C.M.); (D.R.); (P.F.); (F.G.)
- InterInstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele, 00166 Rome, Italy
| | - Carlo Genovese
- Department of Medicine and Surgery, “Kore” University of Enna, Contrada Santa Panasia, 94100 Enna, Italy;
- Nacture S.r.l, Spin-Off University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
27
|
Kapor S, Radojković M, Santibanez JF. Myeloid-derived suppressor cells: Implication in myeloid malignancies and immunotherapy. Acta Histochem 2024; 126:152183. [PMID: 39029317 DOI: 10.1016/j.acthis.2024.152183] [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: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
Myeloid malignancies stem from a modified hematopoietic stem cell and predominantly include acute myeloid leukemia, myelodysplastic neoplasms, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid-derived suppressor cells (MDSCs) exhibit immunoregulatory properties by governing the innate and adaptive immune systems, creating a permissive and supportive environment for neoplasm growth. This review examines the key characteristics of MDSCs in myeloid malignancies, highlighting that an increased MDSC count corresponds to heightened immunosuppressive capabilities, fostering an immune-tolerant neoplasm microenvironment. Also, this review analyzes and describes the potential of combined cancer therapies, focusing on targeting MDSC generation, expansion, and their inherent immunosuppressive activities to enhance the efficacy of current cancer immunotherapies. A comprehensive understanding of the implications of myeloid malignancies may enhance the exploration of immunotherapeutic strategies for their potential application.
Collapse
Affiliation(s)
- Suncica Kapor
- Department of Hematology, Clinical, and Hospital Center "Dr. Dragiša Mišović-Dedinje,", Heroja Milana Tepića 1, Belgrade 11020, Serbia
| | - Milica Radojković
- Department of Hematology, Clinical, and Hospital Center "Dr. Dragiša Mišović-Dedinje,", Heroja Milana Tepića 1, Belgrade 11020, Serbia; Faculty of Medicine, University of Belgrade, Dr. Subotića Starijeg 8, Belgrade 11000, Serbia
| | - Juan F Santibanez
- Molecular Oncology group, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, POB 102, Belgrade 11129, Serbia; Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, General Gana 1780, Santiago 8370854, Chile.
| |
Collapse
|
28
|
Lana JF, de Brito GC, Kruel A, Brito B, Santos GS, Caliari C, Salamanna F, Sartori M, Barbanti Brodano G, Costa FR, Jeyaraman M, Dallo I, Bernaldez P, Purita J, de Andrade MAP, Everts PA. Evolution and Innovations in Bone Marrow Cellular Therapy for Musculoskeletal Disorders: Tracing the Historical Trajectory and Contemporary Advances. Bioengineering (Basel) 2024; 11:979. [PMID: 39451354 PMCID: PMC11504458 DOI: 10.3390/bioengineering11100979] [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: 08/12/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Bone marrow cellular therapy has undergone a remarkable evolution, significantly impacting the treatment of musculoskeletal disorders. This review traces the historical trajectory from early mythological references to contemporary scientific advancements. The groundbreaking work of Friedenstein in 1968, identifying fibroblast colony-forming cells in bone marrow, laid the foundation for future studies. Caplan's subsequent identification of mesenchymal stem cells (MSCs) in 1991 highlighted their differentiation potential and immunomodulatory properties, establishing them as key players in regenerative medicine. Contemporary research has focused on refining techniques for isolating and applying bone marrow-derived MSCs. These cells have shown promise in treating conditions like osteonecrosis, osteoarthritis, and tendon injuries thanks to their ability to promote tissue repair, modulate immune responses, and enhance angiogenesis. Clinical studies have demonstrated significant improvements in pain relief, functional recovery, and tissue regeneration. Innovations such as the ACH classification system and advancements in bone marrow aspiration methods have standardized practices, improving the consistency and efficacy of these therapies. Recent clinical trials have validated the therapeutic potential of bone marrow-derived products, highlighting their advantages in both surgical and non-surgical applications. Studies have shown that MSCs can reduce inflammation, support bone healing, and enhance cartilage repair. However, challenges remain, including the need for rigorous characterization of cell populations and standardized reporting in clinical trials. Addressing these issues is crucial for advancing the field and ensuring the reliable application of these therapies. Looking ahead, future research should focus on integrating bone marrow-derived products with other regenerative techniques and exploring non-surgical interventions. The continued innovation and refinement of these therapies hold promise for revolutionizing the treatment of musculoskeletal disorders, offering improved patient outcomes, and advancing the boundaries of medical science.
Collapse
Affiliation(s)
- José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (G.C.d.B.); (A.K.); (B.B.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (I.D.); (J.P.); (P.A.E.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
- Clinical Research, Anna Vitória Lana Institute (IAVL), Indaiatuba 13334-170, SP, Brazil
- Medical School, Jaguariúna University Center (UniFAJ), Jaguariúna 13820-000, SP, Brazil
| | - Gabriela Caponero de Brito
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (G.C.d.B.); (A.K.); (B.B.)
| | - André Kruel
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (G.C.d.B.); (A.K.); (B.B.)
| | - Benjamim Brito
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (G.C.d.B.); (A.K.); (B.B.)
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (G.C.d.B.); (A.K.); (B.B.)
| | - Carolina Caliari
- Cell Therapy, In Situ Terapia Celular, Ribeirão Preto 14056-680, SP, Brazil;
| | - Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS Instituto Ortopedizo Rizzoli, 40136 Bologna, Italy; (F.S.); (M.S.)
| | - Maria Sartori
- Surgical Sciences and Technologies, IRCCS Instituto Ortopedizo Rizzoli, 40136 Bologna, Italy; (F.S.); (M.S.)
| | | | - Fábio Ramos Costa
- Department of Orthopaedics, FC Sports Traumatology, Salvador 40296-210, BA, Brazil;
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India;
- Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Clinical Research Scientist, Virginia Tech India, Chennai 600095, Tamil Nadu, India
| | - Ignácio Dallo
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (I.D.); (J.P.); (P.A.E.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
- Orthopedics, SportMe Medical Center, 41013 Seville, Spain;
| | | | - Joseph Purita
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (I.D.); (J.P.); (P.A.E.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| | | | - Peter Albert Everts
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (I.D.); (J.P.); (P.A.E.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
- Gulf Coast Biologics, Fort Myers, FL 33916, USA
| |
Collapse
|
29
|
Kegyes D, Thiagarajan PS, Ghiaur G. MRD in Acute Leukemias: Lessons Learned from Acute Promyelocytic Leukemia. Cancers (Basel) 2024; 16:3208. [PMID: 39335179 PMCID: PMC11430625 DOI: 10.3390/cancers16183208] [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: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Introduction: Advances in molecular biology, polymerase chain reaction (PCR), and next-generation sequencing (NGS) have transformed the concept of minimal residual disease (MRD) from a philosophical idea into a measurable reality. Current Treatment Paradigms and Lessons Learned from APL: Acute promyelocytic leukemia (APL) leads the way in this transformation, initially using PCR to detect MRD in patients in remission, and more recently, aiming to eliminate it entirely with modern treatment strategies. Along the way, we have gained valuable insights that, when applied to other forms of acute leukemia, hold the potential to significantly improve the outcomes of these challenging diseases. Does the BM Microenvironment Play a Role in MRD?: In this review, we explore the current use of MRD in the management of acute leukemia and delve into the biological processes that contribute to MRD persistence, including its overlap with leukemia stem cells and the role of the bone marrow microenvironment.
Collapse
Affiliation(s)
- David Kegyes
- MedFuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
- The Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Gabriel Ghiaur
- The Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
30
|
Winkler T, Geissler S, Maleitzke T, Perka C, Duda GN, Hildebrandt A. Advanced therapies in orthopaedics. EFORT Open Rev 2024; 9:837-844. [PMID: 39222330 PMCID: PMC11457816 DOI: 10.1530/eor-24-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Advanced therapies are expected to play a crucial role in supporting repair after injury, halting the degeneration of musculoskeletal tissue to enable and promote physical activity. Despite advancements, the progress in developing advanced therapies in orthopaedics lags behind specialties like oncology, since innovative regenerative treatment strategies fall short of their expectations in musculoskeletal clinical trials. Researchers should focus on understanding the mechanism of action behind the investigated target before conducting clinical trials. Strategic research networks are needed that not only enhance scientific exchange among like-minded researchers but need to include early on commercial views, companies and venture perspectives, regulatory insights and reimbursement perspectives. Only in such collaborations essential roadblocks towards clinical trials and go-to-patients be overcome.
Collapse
Affiliation(s)
- Tobias Winkler
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Tazio Maleitzke
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Trauma Orthopaedic Research Copenhagen Hvidovre (TORCH), Department of Orthopaedic Surgery, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Perka
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
| | - Georg N Duda
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Hildebrandt
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| |
Collapse
|
31
|
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
|
32
|
Park N, Kim KS, Park CG, Jung HD, Park W, Na K. Adipose-derived stem cell-based anti-inflammatory paracrine factor regulation for the treatment of inflammatory bowel disease. J Control Release 2024; 374:384-399. [PMID: 39173953 DOI: 10.1016/j.jconrel.2024.08.027] [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: 05/24/2024] [Revised: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Stem cell-based therapies offer promising avenues for treating inflammatory diseases owing to their immunomodulatory properties. However, challenges persist regarding their survival and efficacy in inflamed tissues. Our study introduces a novel approach by engineering adipose-derived stem cells (ADSCs) to enhance their viability in inflammatory environments and boost the secretion of paracrine factors for treating inflammatory bowel disease (IBD). An arginine-glycine-aspartate peptide-poly (ethylene glycol)-chlorin e6 conjugate (RPC) was synthesized and coupled with ADSCs, resulting in RPC-labeled ADSCs (ARPC). This conjugation strategy employed RGD-integrin interaction to shield stem cells and allowed visualization and tracking using chlorin e6. The engineered ARPC demonstrated enhanced viability and secretion of paracrine factors upon light irradiation, regulating the inflammatory microenvironment. RNA-sequencing analysis unveiled pathways favoring angiogenesis, DNA repair, and exosome secretion in ARPC(+) while downregulating inflammatory pathways. In in vivo models of acute and chronic IBD, ARPC(+) treatment led to reduced inflammation, preserved colon structure, and increased populations of regulatory T cells, highlighting its therapeutic potential. ARPC(+) selectively homed to inflammatory sites, demonstrating its targeted effect. Overall, ARPC(+) exhibits promise as an effective and safe therapeutic strategy for managing inflammatory diseases like IBD by modulating immune responses and creating an anti-inflammatory microenvironment.
Collapse
Affiliation(s)
- Naeun Park
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyoung Sub Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi 16419, Republic of Korea
| | - Hyun-Do Jung
- Division of Materials Science and Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi 16419, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea.
| |
Collapse
|
33
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
34
|
Hu S, Liang Y, Pan X. Exosomes: A promising new strategy for treating osteoporosis in the future. J Drug Deliv Sci Technol 2024; 97:105571. [DOI: 10.1016/j.jddst.2024.105571] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
35
|
Sindeeva OA, Demina PA, Kozyreva ZV, Terentyeva DA, Gusliakova OI, Muslimov AR, Sukhorukov GB. Single Mesenchymal Stromal Cell Migration Tracking into Glioblastoma Using Photoconvertible Vesicles. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1215. [PMID: 39057891 PMCID: PMC11279842 DOI: 10.3390/nano14141215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Reliable cell labeling and tracking techniques are imperative for elucidating the intricate and ambiguous interactions between mesenchymal stromal cells (MSCs) and tumors. Here, we explore fluorescent photoconvertible nanoengineered vesicles to study mMSC migration in brain tumors. These 3 μm sized vesicles made of carbon nanoparticles, Rhodamine B (RhB), and polyelectrolytes are readily internalized by cells. The dye undergoes photoconversion under 561 nm laser exposure with a fluorescence blue shift upon demand. The optimal laser irradiation duration for photoconversion was 0.4 ms, which provided a maximal blue shift of the fluorescent signal label without excessive laser exposure on cells. Vesicles modified with an extra polymer layer demonstrated enhanced intracellular uptake without remarkable effects on cell viability, motility, or proliferation. The optimal ratio of 20 vesicles per mMSC was determined. Moreover, the migration of individual mMSCs within 2D and 3D glioblastoma cell (EPNT-5) colonies over 2 days and in vivo tumor settings over 7 days were traced. Our study provides a robust nanocomposite platform for investigating MSC-tumor dynamics and offers insights into envisaged therapeutic strategies. Photoconvertible vesicles also present an indispensable tool for studying complex fundamental processes of cell-cell interactions for a wide range of problems in biomedicine.
Collapse
Affiliation(s)
- Olga A. Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
| | - Polina A. Demina
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia;
| | - Zhanna V. Kozyreva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
| | - Daria A. Terentyeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
| | - Olga I. Gusliakova
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia;
| | - Albert R. Muslimov
- Center for Molecular and Cell Technologies, Saint Petersburg State Chemical and Pharmaceutical University, 14 Professora Popova Str., lit. A, 197022 St. Petersburg, Russia;
| | - Gleb B. Sukhorukov
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
- Life Improvement by Future Technology (LIFT) Center, 121205 Moscow, Russia
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
36
|
Nie DQ, Yan GX, Wang ZY, Yan X, Yu GM, Gao JL, Liu D, Li HB. Combination treatment with interferon-γ may be a potential strategy to improve the efficacy of cytotherapy for rheumatoid arthritis: A network meta-analysis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:29. [PMID: 39239074 PMCID: PMC11376714 DOI: 10.4103/jrms.jrms_697_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 03/21/2022] [Accepted: 12/07/2022] [Indexed: 09/07/2024]
Abstract
Background Mesenchymal stem cells (MSCs) are considered a promising therapeutic strategy for rheumatoid arthritis (RA), but the current clinical results are varied. This study is to analyze the therapeutic effect of cell-based strategies on RA. Materials and Methods The searches were performed with public databases from inception to June 17, 2021. Randomized controlled trials researching cell-based therapies in RA patients were included. Results Eight studies, including 480 patients, were included in the analysis. The results showed that compared to the control, MSC treatment significantly reduced the disease activity score (DAS) at the second standardized mean difference (SMD): -0.70; 95% confidence interval (CI): -1.25, -0.15; P = 0.01) and 3rd month (SMD: -1.47; 95% CI: -2.77, -0.18; P < 0.01) and significantly reduced the rheumatoid factor (RF) level at the first (SMD: -0.38; 95% CI: -0.72, -0.05; P = 0.03) and 6th months (SMD: -0.81; 95% CI: -1.32, -0.31; P < 0.01). In the network meta-analysis, MSCs combined with interferon-γ (MSC_IFN) had a significant effect on increasing the American college of rheumatology criteria (ACR) 20, ACR50, and DAS <3.2 populations, had a significant effect on reducing the DAS, and decreased the RF level for a long period. Conclusion MSCs could relieve the DAS of RA patients in the short term and reduce the level of RF. MSC_IFN showed a more obvious effect, which could significantly improve the results of ACR20, ACR50, and DAS <3.2 and reduce the DAS and RF levels.
Collapse
Affiliation(s)
- Da-Qing Nie
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Gui-Xiu Yan
- The Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zheng-Yi Wang
- Department of College of Nursing and Health Science, Nanfang Medical College of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xue Yan
- Department of Rheumatism, The Third Clinical Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Gui-Mei Yu
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jin-Liang Gao
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Di Liu
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Bo Li
- Department of Rheumatism, The Third Clinical Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
37
|
Rahmani A, Soleymani A, Almukhtar M, Behzad Moghadam K, Vaziri Z, Hosein Tabar Kashi A, Adabi Firoozjah R, Jafari Tadi M, Zolfaghari Dehkharghani M, Valadi H, Moghadamnia AA, Gasser RB, Rostami A. Exosomes, and the potential for exosome-based interventions against COVID-19. Rev Med Virol 2024; 34:e2562. [PMID: 38924213 DOI: 10.1002/rmv.2562] [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: 09/08/2023] [Revised: 05/17/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
Since late 2019, the world has been devastated by the coronavirus disease 2019 (COVID-19) induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with more than 760 million people affected and ∼seven million deaths reported. Although effective treatments for COVID-19 are currently limited, there has been a strong focus on developing new therapeutic approaches to address the morbidity and mortality linked to this disease. An approach that is currently being investigated is the use of exosome-based therapies. Exosomes are small, extracellular vesicles that play a role in many clinical diseases, including viral infections, infected cells release exosomes that can transmit viral components, such as miRNAs and proteins, and can also include receptors for viruses that facilitate viral entry into recipient cells. SARS-CoV-2 has the ability to impact the formation, secretion, and release of exosomes, thereby potentially facilitating or intensifying the transmission of the virus among cells, tissues and individuals. Therefore, designing synthetic exosomes that carry immunomodulatory cargo and antiviral compounds are proposed to be a promising strategy for the treatment of COVID-19 and other viral diseases. Moreover, exosomes generated from mesenchymal stem cells (MSC) might be employed as cell-free therapeutic agents, as MSC-derived exosomes can diminish the cytokine storm and reverse the suppression of host anti-viral defences associated with COVID-19, and boost the repair of lung damage linked to mitochondrial activity. The present article discusses the significance and roles of exosomes in COVID-19, and explores potential future applications of exosomes in combating this disease. Despite the challenges posed by COVID-19, exosome-based therapies could represent a promising avenue for improving patient outcomes and reducing the impact of this disease.
Collapse
Affiliation(s)
- Abolfazl Rahmani
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Soleymani
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Kimia Behzad Moghadam
- Independent Researcher, Former University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Zahra Vaziri
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Hosein Tabar Kashi
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Reza Adabi Firoozjah
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mehrdad Jafari Tadi
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Maryam Zolfaghari Dehkharghani
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas (UNLV), Las Vegas, Nevada, USA
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ali Akbar Moghadamnia
- Department of Pharmacology and Toxicology, Babol University of Medical Sciences, Babol, Iran
- Pharmaceutical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Robin B Gasser
- Department of Veterinary Biosciences, Faculty of Science, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria, Australia
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
38
|
Tragoonlugkana P, Pruksapong C, Ontong P, Kamprom W, Supokawej A. Fibronectin and vitronectin alleviate adipose-derived stem cells senescence during long-term culture through the AKT/MDM2/P53 pathway. Sci Rep 2024; 14:14242. [PMID: 38902430 PMCID: PMC11189918 DOI: 10.1038/s41598-024-65339-z] [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: 02/19/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
Cellular senescence plays a role in the development of aging-associated degenerative diseases. Cell therapy is recognized as a candidate treatment for degenerative diseases. To achieve the goal of cell therapy, the quality and good characteristics of cells are concerned. Cell expansion relies on two-dimensional culture, which leads to replicative senescence of expanded cells. This study aimed to investigate the effect of cell culture surface modification using fibronectin (FN) and vitronectin (VN) in adipose-derived stem cells (ADSCs) during long-term expansion. Our results showed that ADSCs cultured in FN and VN coatings significantly enhanced adhesion, proliferation, and slow progression of cellular senescence as indicated by lower SA-β-gal activities and decreased expression levels of genes including p16, p21, and p53. The upregulation of integrin α5 and αv genes influences phosphatidylinositol 4,5-bisphosphate 3-kinase (PI3K), and AKT proteins. FN and VN coatings upregulated AKT and MDM2 leading to p53 degradation. Additionally, MDM2 inhibition by Nutlin-3a markedly elevated p53 and p21 expression, increased cellular senescence, and induced the expression of inflammatory molecules including HMGB1 and IL-6. The understanding of FN and VN coating surface influencing ADSCs, especially senescence characteristics, offers a promising and practical point for the cultivation of ADSCs for future use in cell-based therapies.
Collapse
Affiliation(s)
- Patcharapa Tragoonlugkana
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Chatchai Pruksapong
- Department of Surgery, Phramongkutklao Hospital and Phramongkutklao College of Medicine, Bangkok, 10400, Thailand
| | - Pawared Ontong
- Department of Community Medical Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Witchayapon Kamprom
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
39
|
Bharadwaj S, Groza Y, Mierzwicka JM, Malý P. Current understanding on TREM-2 molecular biology and physiopathological functions. Int Immunopharmacol 2024; 134:112042. [PMID: 38703564 DOI: 10.1016/j.intimp.2024.112042] [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: 02/08/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 05/06/2024]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM-2), a glycosylated receptor belonging to the immunoglobin superfamily and especially expressed in the myeloid cell lineage, is frequently explained as a reminiscent receptor for both adaptive and innate immunity regulation. TREM-2 is also acknowledged to influence NK cell differentiation via the PI3K and PLCγ signaling pathways, as well as the partial activation or direct inhibition of T cells. Additionally, TREM-2 overexpression is substantially linked to cell-specific functions, such as enhanced phagocytosis, reduced toll-like receptor (TLR)-mediated inflammatory cytokine production, increased transcription of anti-inflammatory cytokines, and reshaped T cell function. Whereas TREM-2-deficient cells exhibit diminished phagocytic function and enhanced proinflammatory cytokines production, proceeding to inflammatory injuries and an immunosuppressive environment for disease progression. Despite the growing literature supporting TREM-2+ cells in various diseases, such as neurodegenerative disorders and cancer, substantial facets of TREM-2-mediated signaling remain inadequately understood relevant to pathophysiology conditions. In this direction, herein, we have summarized the current knowledge on TREM-2 biology and cell-specific TREM-2 expression, particularly in the modulation of pivotal TREM-2-dependent functions under physiopathological conditions. Furthermore, molecular regulation and generic biological relevance of TREM-2 are also discussed, which might provide an alternative approach for preventing or reducing TREM-2-associated deformities. At last, we discussed the TREM-2 function in supporting an immunosuppressive cancer environment and as a potential drug target for cancer immunotherapy. Hence, summarized knowledge of TREM-2 might provide a window to overcome challenges in clinically effective therapies for TREM-2-induced diseases in humans.
Collapse
Affiliation(s)
- Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic.
| | - Yaroslava Groza
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Joanna M Mierzwicka
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic.
| |
Collapse
|
40
|
Sadeghi S, Nimtz L, Niebergall-Roth E, Norrick A, Hägele S, Vollmer L, Esterlechner J, Frank MH, Ganss C, Scharffetter-Kochanek K, Kluth MA. Potency assay to predict the anti-inflammatory capacity of a cell therapy product for macrophage-driven diseases: overcoming the challenges of assay development and validation. Cytotherapy 2024; 26:512-523. [PMID: 38441512 PMCID: PMC11065629 DOI: 10.1016/j.jcyt.2024.02.004] [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: 09/13/2023] [Revised: 01/22/2024] [Accepted: 02/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Given the high level of product complexity and limited regulatory guidance, designing and implementing appropriate potency assays is often the most challenging part of establishing a quality control testing matrix for a cell-based medicinal product. Among the most elusive tasks are the selection of suitable read-out parameters, the development of assay designs that most closely model the pathophysiological conditions, and the validation of the methods. Here we describe these challenges and how they were addressed in developing an assay that measures the anti-inflammatory potency of mesenchymal stromal cells (MSCs) in an M1 macrophage-dominated inflammatory environment. METHODS An in vitro inflammation model was established by coculturing skin-derived ABCB5+ MSCs with THP-1 monocyte-derived M1-polarized macrophages. Readout was the amount of interleukin 1 receptor antagonist (IL-1RA) secreted by the MSCs in the coculture, measured by an enzyme-linked immunosorbent assay. RESULTS IL-1RA was quantified with guideline-concordant selectivity, accuracy and precision over a relevant concentration range. Consistent induction of the macrophage markers CD36 and CD80 indicated successful macrophage differentiation and M1 polarization of THP-1 cells, which was functionally confirmed by release of proinflammatory tumor necrosis factor α. Testing a wide range of MSC/macrophage ratios revealed the optimal ratio for near-maximal stimulation of MSCs to secrete IL-1RA, providing absolute maximum levels per individual MSC that can be used for future comparison with clinical efficacy. Batch release testing of 71 consecutively manufactured MSC batches showed a low overall failure rate and a high comparability between donors. CONCLUSIONS We describe the systematic development and validation of a therapeutically relevant, straightforward, robust and reproducible potency assay to measure the immunomodulatory capacity of MSCs in M1 macrophage-driven inflammation. The insights into the challenges and how they were addressed may also be helpful to developers of potency assays related to other cellular functions and clinical indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA; Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | | | | | | |
Collapse
|
41
|
He J, Liu B, Du X, Wei Y, Kong D, Feng B, Guo R, Asiamah EA, Griffin MD, Hynes SO, Shen S, Liu Y, Cui H, Ma J, O'Brien T. Amelioration of diabetic nephropathy in mice by a single intravenous injection of human mesenchymal stromal cells at early and later disease stages is associated with restoration of autophagy. Stem Cell Res Ther 2024; 15:66. [PMID: 38443965 PMCID: PMC10916232 DOI: 10.1186/s13287-024-03647-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND AND AIMS Mesenchymal stromal cells (MSCs) a potentially effective disease-modulating therapy for diabetic nephropathy (DN) but their clinical translation has been hampered by incomplete understanding of the optimal timing of administration and in vivo mechanisms of action. This study aimed to elucidate the reno-protective potency and associated mechanisms of single intravenous injections of human umbilical cord-derived MSCs (hUC-MSCs) following shorter and longer durations of diabetes. METHODS A streptozotocin (STZ)-induced model of diabetes and DN was established in C57BL/6 mice. In groups of diabetic animals, human (h)UC-MSCs or vehicle were injected intravenously at 8 or 16 weeks after STZ along with vehicle-injected non-diabetic animals. Diabetes-related kidney abnormalities was analyzed 2 weeks later by urine and serum biochemical assays, histology, transmission electron microscopy and immunohistochemistry. Serum concentrations of pro-inflammatory and pro-fibrotic cytokines were quantified by ELISA. The expression of autophagy-related proteins within the renal cortices was investigated by immunoblotting. Bio-distribution of hUC-MSCs in kidney and other organs was evaluated in diabetic mice by injection of fluorescent-labelled cells. RESULTS Compared to non-diabetic controls, diabetic mice had increases in urine albumin creatinine ratio (uACR), mesangial matrix deposition, podocyte foot process effacement, glomerular basement membrane thickening and interstitial fibrosis as well as reduced podocyte numbers at both 10 and 18 weeks after STZ. Early (8 weeks) hUC-MSC injection was associated with reduced uACR and improvements in multiple glomerular and renal interstitial abnormalities as well as reduced serum IL-6, TNF-α, and TGF-β1 compared to vehicle-injected animals. Later (16 weeks) hUC-MSC injection also resulted in reduction of diabetes-associated renal abnormalities and serum TGF-β1 but not of serum IL-6 and TNF-α. At both time-points, the kidneys of vehicle-injected diabetic mice had higher ratio of p-mTOR to mTOR, increased abundance of p62, lower abundance of ULK1 and Atg12, and reduced ratio of LC3B to LC3A compared to non-diabetic animals, consistent with diabetes-associated suppression of autophagy. These changes were largely reversed in the kidneys of hUC-MSC-injected mice. In contrast, neither early nor later hUC-MSC injection had effects on blood glucose and body weight of diabetic animals. Small numbers of CM-Dil-labeled hUC-MSCs remained detectable in kidneys, lungs and liver of diabetic mice at 14 days after intravenous injection. CONCLUSIONS Single intravenous injections of hUC-MSCs ameliorated glomerular abnormalities and interstitial fibrosis in a mouse model of STZ-induced diabetes without affecting hyperglycemia, whether administered at relatively short or longer duration of diabetes. At both time-points, the reno-protective effects of hUC-MSCs were associated with reduced circulating TGF-β1 and restoration of intra-renal autophagy.
Collapse
Affiliation(s)
- Jingjing He
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Boxin Liu
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Xiaofeng Du
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Yan Wei
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Desheng Kong
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Baofeng Feng
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Human Anatomy Department, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
| | - Ruiyun Guo
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
| | - Ernest Amponsah Asiamah
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Department of Forensic Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, PMB UCC, Cape Coast, Ghana
| | - Matthew D Griffin
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Sean O Hynes
- Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| | - Sanbing Shen
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Yan Liu
- Department of Endocrinology, Hebei Medical University Third Affiliated Hospital, Shijiazhuang, 050051, Hebei, China
| | - Huixian Cui
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China
- Human Anatomy Department, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
| | - Jun Ma
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, 050017, Hebei Province, China.
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China.
- Hebei International Joint Research Center for Stem Cell and Regenerative Medicine, Shijiazhuang, 050017, Hebei Province, China.
- Human Anatomy Department, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China.
| | - Timothy O'Brien
- Hebei Medical University-University of Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China.
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland.
| |
Collapse
|
42
|
Cavaliere F, Allegri M, Apan A, Brazzi L, Carassiti M, Cohen E, DI Marco P, Langeron O, Rossi M, Spieth P, Turnbull D, Weber F. A year in review in Minerva Anestesiologica 2023: anesthesia, analgesia, and perioperative medicine. Minerva Anestesiol 2024; 90:222-234. [PMID: 38535972 DOI: 10.23736/s0375-9393.24.18067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Affiliation(s)
- Franco Cavaliere
- IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy -
| | - Massimo Allegri
- Lemanic Center of Analgesia and Neuromodulation EHC, Morges, Switzerland
| | - Alparslan Apan
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, University of Giresun, Giresun, Türkiye
| | - Luca Brazzi
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Massimiliano Carassiti
- Unit of Anesthesia, Intensive Care and Pain Management, Campus Bio-Medico University Hospital, Rome, Italy
| | - Edmond Cohen
- Department of Anesthesiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pierangelo DI Marco
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic, and Geriatric Sciences, Faculty of Medicine, Sapienza University, Rome, Italy
| | - Olivier Langeron
- Department of Anesthesia and Intensive Care, Henri Mondor University Hospital, Assistance Publique - Hôpitaux de Paris (APHP), University Paris-Est Créteil (UPEC), Paris, France
| | - Marco Rossi
- IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Peter Spieth
- Department of Anesthesiology and Critical Care Medicine, University Hospital of Dresden, Dresden, Germany
| | - David Turnbull
- Department of Anesthetics and Neuro Critical Care, Royal Hallamshire Hospital, Sheffield, UK
| | - Frank Weber
- Department of Anesthesiology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
43
|
Vukotić M, Kapor S, Simon F, Cokic V, Santibanez JF. Mesenchymal stromal cells in myeloid malignancies: Immunotherapeutic opportunities. Heliyon 2024; 10:e25081. [PMID: 38314300 PMCID: PMC10837636 DOI: 10.1016/j.heliyon.2024.e25081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.
Collapse
Affiliation(s)
- Milica Vukotić
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje,” University of Belgrade, Serbia
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile
| | - Vladan Cokic
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Juan F. Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
44
|
Śmieszek A, Marcinkowska K, Małas Z, Sikora M, Kępska M, Nowakowska BA, Deperas M, Smyk M, Rodriguez-Galindo C, Raciborska A. Identification and characterization of stromal-like cells with CD207 +/low CD1a +/low phenotype derived from histiocytic lesions - a perspective in vitro model for drug testing. BMC Cancer 2024; 24:105. [PMID: 38342891 PMCID: PMC10860276 DOI: 10.1186/s12885-023-11807-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 12/28/2023] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND Histiocytoses are rare disorders manifested by increased proliferation of pathogenic myeloid cells sharing histological features with macrophages or dendritic cells and accumulating in various organs, i.a., bone and skin. Pre-clinical in vitro models that could be used to determine molecular pathways of the disease are limited, hence research on histiocytoses is challenging. The current study compares cytophysiological features of progenitor, stromal-like cells derived from histiocytic lesions (sl-pHCs) of three pediatric patients with different histiocytoses types and outcomes. The characterized cells may find potential applications in drug testing. METHODS Molecular phenotype of the cells, i.e. expression of CD1a and CD207 (langerin), was determined using flow cytometry. Cytogenetic analysis included GTG-banded metaphases and microarray (aCGH) evaluation. Furthermore, the morphology and ultrastructure of cells were evaluated using a confocal and scanning electron microscope. The microphotographs from the confocal imaging were used to reconstruct the mitochondrial network and its morphology. Basic cytophysiological parameters, such as viability, mitochondrial activity, and proliferation, were analyzed using multiple cellular assays, including Annexin V/7-AAD staining, mitopotential analysis, BrdU test, clonogenicity analysis, and distribution of cells within the cell cycle. Biomarkers potentially associated with histiocytoses progression were determined using RT-qPCR at mRNA, miRNA and lncRNA levels. Intracellular accumulation of histiocytosis-specific proteins was detected with Western blot. Cytotoxicyty and IC50 of vemurafenib and trametinib were determined with MTS assay. RESULTS Obtained cellular models, i.e. RAB-1, HAN-1, and CHR-1, are heterogenic in terms of molecular phenotype and morphology. The cells express CD1a/CD207 markers characteristic for dendritic cells, but also show intracellular accumulation of markers characteristic for cells of mesenchymal origin, i.e. vimentin (VIM) and osteopontin (OPN). In subsequent cultures, cells remain viable and metabolically active, and the mitochondrial network is well developed, with some distinctive morphotypes noted in each cell line. Cell-specific transcriptome profile was noted, providing information on potential new biomarkers (non-coding RNAs) with diagnostic and prognostic features. The cells showed different sensitivity to vemurafenib and trametinib. CONCLUSION Obtained and characterized cellular models of stromal-like cells derived from histiocytic lesions can be used for studies on histiocytosis biology and drug testing.
Collapse
Affiliation(s)
- Agnieszka Śmieszek
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375, Wroclaw, Poland.
| | - Klaudia Marcinkowska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Zofia Małas
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Martyna Kępska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Beata A Nowakowska
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Marta Deperas
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Marta Smyk
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | | | - Anna Raciborska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland.
| |
Collapse
|
45
|
Qiu Y, Li C, Sheng S. Efficacy and safety of stem cell therapy for Crohn's disease: a meta-analysis of randomized controlled trials. Stem Cell Res Ther 2024; 15:28. [PMID: 38303054 PMCID: PMC10835827 DOI: 10.1186/s13287-024-03637-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
PURPOSE Small-scale clinical trials have provided evidence suggesting the effectiveness of stem-cell therapy (SCT) for patients diagnosed with Crohn's disease (CD). The objective of the research was to systematically assess the effectiveness and safety of SCT for individuals diagnosed with CD through a comprehensive review and meta-analysis. METHODS A search was conducted in Medline (PubMed), CENTER (Cochrane Library), and Embase (Ovid) to find randomized controlled trials (RCTs) that assessed the impact of SCT on the occurrence of clinical remission (CR) and severe adverse events (SAE) among patients diagnosed with CD. The Cochrane Q test and estimation of I2 were used to assess heterogeneity among studies. After incorporating heterogeneity, a random-effects model was employed for data pooling. RESULTS Overall, 12 RCTs involving 632 adult patients with medically refractory CD or CD-related fistula were included. In comparison with placebo or no treatment, SCT showed a greater likelihood of CR (odds ratio [OR] 2.08, 95% CI 1.39-3.12, p < 0.001) without any notable heterogeneity (I2 = 0%). Consistent results were observed in subgroup analyses based on study design, patient diagnosis, source and type of stem cells, and follow-up durations, with all p-values for subgroup analyses being greater than 0.05. The occurrence of SAE was similar among patients assigned to SCT and the placebo/no treatment cohorts (OR 0.70, 95% CI 0.37-1.33, p = 0.28; I2 = 0%). CONCLUSIONS For patients with medically refractory CD or CD-related fistula, SCT may be an alternatively effective and safe treatment.
Collapse
Affiliation(s)
- Yunfeng Qiu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Shihou Sheng
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033, China.
| |
Collapse
|
46
|
Towers R, Trombello L, Fusenig M, Tunger A, Baumann AL, Savoldelli R, Wehner R, Fasslrinner F, Arndt C, Dazzi F, Von Bonin M, Feldmann A, Bachmann MP, Wobus M, Schmitz M, Bornhäuser M. Bone marrow-derived mesenchymal stromal cells obstruct AML-targeting CD8 + clonal effector and CAR T-cell function while promoting a senescence-associated phenotype. Cancer Immunol Immunother 2024; 73:8. [PMID: 38231344 PMCID: PMC10794426 DOI: 10.1007/s00262-023-03594-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/03/2023] [Indexed: 01/18/2024]
Abstract
Bone marrow mesenchymal stromal cells (MSCs) have been described as potent regulators of T-cell function, though whether they could impede the effectiveness of immunotherapy against acute myeloid leukemia (AML) is still under investigation. We examine whether they could interfere with the activity of leukemia-specific clonal cytotoxic T-lymphocytes (CTLs) and chimeric antigen receptor (CAR) T cells, as well as whether the immunomodulatory properties of MSCs could be associated with the induction of T-cell senescence. Co-cultures of leukemia-associated Wilm's tumor protein 1 (WT1) and tyrosine-protein kinase transmembrane receptor 1 (ROR1)-reactive CTLs and of CD123-redirected switchable CAR T cells were prepared in the presence of MSCs and assessed for cytotoxic potential, cytokine secretion, and expansion. T-cell senescence within functional memory sub-compartments was investigated for the senescence-associated phenotype CD28-CD57+ using unmodified peripheral blood mononuclear cells. We describe inhibition of expansion of AML-redirected switchable CAR T cells by MSCs via indoleamine 2,3-dioxygenase 1 (IDO-1) activity, as well as reduction of interferon gamma (IFNγ) and interleukin-2 (IL-2) release. In addition, MSCs interfered with the secretory potential of leukemia-associated WT1- and ROR1-targeting CTL clones, inhibiting the release of IFNγ, tumor necrosis factor alpha, and IL-2. Abrogated T cells were shown to retain their cytolytic activity. Moreover, we demonstrate induction of a CD28loCD27loCD57+KLRG1+ senescent T-cell phenotype by MSCs. In summary, we show that MSCs are potent modulators of anti-leukemic T cells, and targeting their modes of action would likely be beneficial in a combinatorial approach with AML-directed immunotherapy.
Collapse
Affiliation(s)
- Russell Towers
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- National Centre for Tumor Disease (NCT/UCC), Fetscherstraße 74, 01307, Dresden, Germany
| | - Lidia Trombello
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà 33, 56127, Pisa, Italy
- University of Pisa, Lungarno Antonio Pacinotti 43, 56126, Pisa, Italy
| | - Maximilian Fusenig
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| | - Antje Tunger
- National Centre for Tumor Disease (NCT/UCC), Fetscherstraße 74, 01307, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Anna-Lena Baumann
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Roberto Savoldelli
- School of Cancer and Pharmaceutical Research, Kings College, London, SE5 9RS, UK
| | - Rebekka Wehner
- National Centre for Tumor Disease (NCT/UCC), Fetscherstraße 74, 01307, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- Partner Site Dresden, and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Frederick Fasslrinner
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Mildred Scheel Early Career Center, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Claudia Arndt
- Department of Radioimmunology, Helmholtz Center Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzener Straße 400, 01328, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Mildred Scheel Early Career Center, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Francesco Dazzi
- School of Cancer and Pharmaceutical Research, Kings College, London, SE5 9RS, UK
| | - Malte Von Bonin
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Helmholtz Center Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzener Straße 400, 01328, Dresden, Germany
| | - Michael P Bachmann
- National Centre for Tumor Disease (NCT/UCC), Fetscherstraße 74, 01307, Dresden, Germany
- Partner Site Dresden, and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Radioimmunology, Helmholtz Center Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzener Straße 400, 01328, Dresden, Germany
| | - Manja Wobus
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Marc Schmitz
- National Centre for Tumor Disease (NCT/UCC), Fetscherstraße 74, 01307, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- Partner Site Dresden, and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Martin Bornhäuser
- Medical Clinic 1 (MK1), University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
- National Centre for Tumor Disease (NCT/UCC), Fetscherstraße 74, 01307, Dresden, Germany.
- School of Cancer and Pharmaceutical Research, Kings College, London, SE5 9RS, UK.
- Partner Site Dresden, and German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
47
|
Kelly CJ, Lindsay SL, Smith RS, Keh S, Cunningham KT, Thümmler K, Maizels RM, Campbell JDM, Barnett SC. Development of Good Manufacturing Practice-Compatible Isolation and Culture Methods for Human Olfactory Mucosa-Derived Mesenchymal Stromal Cells. Int J Mol Sci 2024; 25:743. [PMID: 38255817 PMCID: PMC10815924 DOI: 10.3390/ijms25020743] [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: 11/23/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Demyelination in the central nervous system (CNS) resulting from injury or disease can cause loss of nerve function and paralysis. Cell therapies intended to promote remyelination of axons are a promising avenue of treatment, with mesenchymal stromal cells (MSCs) a prominent candidate. We have previously demonstrated that MSCs derived from human olfactory mucosa (hOM-MSCs) promote myelination to a greater extent than bone marrow-derived MSCs (hBM-MSCs). However, hOM-MSCs were developed using methods and materials that were not good manufacturing practice (GMP)-compliant. Before considering these cells for clinical use, it is necessary to develop a method for their isolation and expansion that is readily adaptable to a GMP-compliant environment. We demonstrate here that hOM-MSCs can be derived without enzymatic tissue digestion or cell sorting and without culture antibiotics. They grow readily in GMP-compliant media and express typical MSC surface markers. They robustly produce CXCL12 (a key secretory factor in promoting myelination) and are pro-myelinating in in vitro rodent CNS cultures. GMP-compliant hOM-MSCs are comparable in this respect to those grown in non-GMP conditions. However, when assessed in an in vivo model of demyelinating disease (experimental autoimmune encephalitis, EAE), they do not significantly improve disease scores compared with controls, indicating further pre-clinical evaluation is necessary before their advancement to clinical trials.
Collapse
Affiliation(s)
- Christopher J. Kelly
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Susan L. Lindsay
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Rebecca Sherrard Smith
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Siew Keh
- New Victoria Hospital, 55 Grange Road, Glasgow G42 9LF, UK
| | - Kyle T. Cunningham
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Katja Thümmler
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - Rick M. Maizels
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| | - John D. M. Campbell
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
- Tissues Cells and Advanced Therapeutics, SNBTS, Jack Copland Centre, Edinburgh EH14 4BE, UK
| | - Susan C. Barnett
- School of Infection and Immunity, 120 University Place, Glasgow G12 8TA, UK; (C.J.K.); (S.L.L.); (R.M.M.)
| |
Collapse
|
48
|
Mao XF, Zhang XQ, Yao ZY, Mao HJ. Advances in mesenchymal stem cells therapy for tendinopathies. Chin J Traumatol 2024; 27:11-17. [PMID: 38052701 PMCID: PMC10859297 DOI: 10.1016/j.cjtee.2023.11.002] [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: 02/23/2023] [Revised: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023] Open
Abstract
Tendinopathies are chronic diseases of an unknown etiology and associated with inflammation. Mesenchymal stem cells (MSCs) have emerged as a viable therapeutic option to combat the pathological progression of tendinopathies, not only because of their potential for multidirectional differentiation and self-renewal, but also their excellent immunomodulatory properties. The immunomodulatory effects of MSCs are increasingly being recognized as playing a crucial role in the treatment of tendinopathies, with MSCs being pivotal in regulating the inflammatory microenvironment by modulating the immune response, ultimately contributing to improved tissue repair. This review will discuss the current knowledge regarding the application of MSCs in tendinopathy treatments through the modulation of the immune response.
Collapse
Affiliation(s)
- Xu-Feng Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China
| | - Xi-Qian Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China
| | - Zhe-Yu Yao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China
| | - Hai-Jiao Mao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang province, China.
| |
Collapse
|
49
|
Burnham AJ, Foppiani EM, Goss KL, Jang-Milligan F, Kamalakar A, Bradley H, Goudy SL, Trochez CM, Dominici M, Daley-Bauer L, Gibson G, Horwitz EM. Differential response of mesenchymal stromal cells (MSCs) to type 1 ex vivo cytokine priming: implications for MSC therapy. Cytotherapy 2023; 25:1277-1284. [PMID: 37815775 DOI: 10.1016/j.jcyt.2023.08.014] [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: 06/01/2022] [Revised: 07/19/2023] [Accepted: 08/30/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are polymorphic, adherent cells with the capability to stimulate tissue regeneration and modulate immunity. MSCs have been broadly investigated for potential therapeutic applications, particularly immunomodulatory properties, wound healing and tissue regeneration. The exact physiologic role of MSCs, however, remains poorly understood, and this gap in knowledge significantly impedes the rational development of therapeutic cells. Here, we considered interferon γ (IFN-γ) and tumor necrosis factor alpha (TNF-α), two cytokines likely encountered physiologically and commonly used in cell manufacturing. For comparison, we studied interleukin-10 (IL-10) (anti-inflammatory) and interleukin-4 (IL-4) (type 2 cytokine). METHODS We directly assessed the effects of these cytokines on bone marrow MSCs by comparing RNA Seq transcriptional profiles. Western blotting and flow cytometry were also used to evaluate effects of cytokine priming. RESULTS The type 1 cytokines (IFN-γ and TNF-α) induced striking changes in gene expression and remarkably different profiles from one another. Importantly, priming MSCs with either of these cytokines did not increase variability among multiple donors beyond what is intrinsic to non-primed MSCs from different donors. IFN-γ-primed MSCs expressed IDO1 and chemokines that recruit activated T cells. In contrast, TNF-α-primed MSCs expressed genes in alternate pathways, namely PGE2 and matrix metalloproteinases synthesis, and chemokines that recruit neutrophils. IL-10 and IL-4 priming had little to no effect. CONCLUSIONS Our data suggest that IFN-γ-primed MSCs may be a more efficacious immunosuppressive therapy aimed at diseases that target T cells (ie, graft-versus-host disease) compared with TNF-α-primed or non-primed MSCs, which may be better suited for therapies in other disease settings. These results contribute to our understanding of MSC bioactivity and suggest rational ex vivo cytokine priming approaches for MSC manufacturing and therapeutic applications.
Collapse
Affiliation(s)
- Andre J Burnham
- Department of Pediatrics, Marcus Center for Pediatric Cellular Therapy, Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Elisabetta M Foppiani
- Department of Pediatrics, Marcus Center for Pediatric Cellular Therapy, Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kyndal L Goss
- Department of Pediatrics, Marcus Center for Pediatric Cellular Therapy, Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA; Division of Biologic and Biomedical Sciences, Laney Graduate School, Emory University Atlanta, Georgia, USA
| | - Fraser Jang-Milligan
- Department of Pediatrics, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Archana Kamalakar
- Department of Otolaryngology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Heath Bradley
- Department of Otolaryngology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Steven L Goudy
- Department of Otolaryngology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Lisa Daley-Bauer
- Department of Pediatrics, Marcus Center for Pediatric Cellular Therapy, Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Edwin M Horwitz
- Department of Pediatrics, Marcus Center for Pediatric Cellular Therapy, Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA; Division of Biologic and Biomedical Sciences, Laney Graduate School, Emory University Atlanta, Georgia, USA.
| |
Collapse
|
50
|
Deptuła M, Zawrzykraj M, Sawicka J, Banach-Kopeć A, Tylingo R, Pikuła M. Application of 3D- printed hydrogels in wound healing and regenerative medicine. Biomed Pharmacother 2023; 167:115416. [PMID: 37683592 DOI: 10.1016/j.biopha.2023.115416] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Hydrogels are three-dimensional polymer networks with hydrophilic properties. The modifiable properties of hydrogels and the structure resembling living tissue allow their versatile application. Therefore, increasing attention is focused on the use of hydrogels as bioinks for three-dimensional (3D) printing in tissue engineering. Bioprinting involves the fabrication of complex structures from several types of materials, cells, and bioactive compounds. Stem cells (SC), such as mesenchymal stromal cells (MSCs) are frequently employed in 3D constructs. SCs have desirable biological properties such as the ability to differentiate into various types of tissue and high proliferative capacity. Encapsulating SCs in 3D hydrogel constructs enhances their reparative abilities and improves the likelihood of reaching target tissues. In addition, created constructs can simulate the tissue environment and mimic biological signals. Importantly, the immunogenicity of scaffolds is minimized through the use of patient-specific cells and the biocompatibility and biodegradability of the employed biopolymers. Regenerative medicine is taking advantage of the aforementioned capabilities in regenerating various tissues- muscle, bones, nerves, heart, skin, and cartilage.
Collapse
Affiliation(s)
- Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Medical University of Gdansk, Poland.
| | | | - Justyna Sawicka
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Poland
| | - Adrianna Banach-Kopeć
- Department of Chemistry, Technology and Biochemistry of Food, Faculty of Chemistry, Gdansk University of Technology, Poland
| | - Robert Tylingo
- Department of Chemistry, Technology and Biochemistry of Food, Faculty of Chemistry, Gdansk University of Technology, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Medical University of Gdansk, Poland
| |
Collapse
|