1
|
Zununi Vahed S, Hejazian SM, Bakari WN, Landon R, Gueguen V, Meddahi-Pellé A, Anagnostou F, Barzegari A, Pavon-Djavid G. Milking mesenchymal stem cells: Updated protocols for cell lysate, secretome, and exosome extraction, and comparative analysis of their therapeutic potential. Methods 2025; 238:40-60. [PMID: 40058715 DOI: 10.1016/j.ymeth.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/21/2025] Open
Abstract
The potential of the cell lysate, secretome, and extracellular vesicles (EVs) of mesenchymal stem cells (MSCs) to modulate the immune response and promote tissue regeneration has positioned them as a promising option for cell-free therapy. Currently, many clinical trials in stem cells-derived EVs and secretome are in progress various diseases and sometimes the results are failing. The major challenge on this roadmap is the lack of a standard extraction method for exosome, secretome, and lysate. The most optimal method for obtaining the secretome of MSCs for clinical utilization involves a comprehensive approach that includes non-destructive collection methods, time optimization, multiple collection rounds, optimization of culture conditions, and quality control measures. Further research and clinical studies are warranted to validate and refine these methods for safe and effective utilization of the MSC exosome, secretome, and lysate in various clinical applications. To address these challenges, it is imperative to establish a standardized and unified methodology to ensure reliable evaluation of these extractions in clinical trials. This review seeks to outline the pros and cons of methods for the preparation of MSCs-derived exosome, and secretome/lysate, and comparative analysis of their therapeutic potential.
Collapse
Affiliation(s)
| | | | - William Ndjidda Bakari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France; Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Rebecca Landon
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Fani Anagnostou
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France.
| |
Collapse
|
2
|
Jiang M, Bu W, Wang X, Ruan J, Shi W, Yu S, Huang L, Xue P, Tang J, Zhao X, Su L, Cheng D. Pulmonary fibrosis: from mechanisms to therapies. J Transl Med 2025; 23:515. [PMID: 40340941 PMCID: PMC12063347 DOI: 10.1186/s12967-025-06514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive interstitial lung disease characterized by excessive deposition of extracellular matrix (ECM) and abnormal fibroblast proliferation, which is mainly caused by air pollution, smoking, aging, occupational exposure, environmental pollutants exposure, and microbial infections. Although antifibrotic agents such as pirfenidone and nintedanib, approved by the United States (US) Food and Drug Administration (FDA), can slow the decline in lung function and disease progression, their side effects and delivery inefficiency limit the overall prognosis of PF. Therefore, there is an urgent need to develop effective therapeutic targets and delivery approaches for PF in clinical settings. This review provides an overview of the pathogenic mechanisms, therapeutic drug targeting signaling pathways, and promising drug delivery strategies for treating PF.
Collapse
Affiliation(s)
- Mengna Jiang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Wenxia Bu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xuehai Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Jialing Ruan
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Weijian Shi
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Siqi Yu
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Lizhen Huang
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Peng Xue
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| | - Liling Su
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China.
| | - Demin Cheng
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| |
Collapse
|
3
|
Rolandsson Enes S, Dzneladze I, Hampton TH, Neff SL, Asarian L, Barua J, Tertel T, Giebel B, Pereyra N, McKenna DH, Hu P, Acton E, Ashare A, Liu KD, Krasnodembskaya AD, English K, Stanton BA, Rocco PRM, Matthay MA, Dos Santos CC, Weiss DJ. Acute respiratory distress vs healthy lung environments differently affect mesenchymal stromal cell extracellular vesicle miRNAs. Cytotherapy 2025; 27:581-596. [PMID: 39945694 DOI: 10.1016/j.jcyt.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 05/24/2025]
Abstract
The acute respiratory distress syndrome (ARDS) inflammatory environment alters mesenchymal stromal cell (MSC) gene and protein expression but effects on microRNA (miRNA) content of MSC-extracellular vesicle (EVs) remain unknown. To assess this, sequencing analysis of EV-miRNAs prepared from human bone marrow-derived MSCs (hMSCs) exposed ex vivo to bronchoalveolar lavage fluid (BALF) from ARDS patients or healthy volunteers (HV) identified a number of differentially expressed miRNAs. Discriminant, differential expression, and functional enrichment analyses identified 14 miRNAs significantly changed following ARDS versus HV BALF exposure. Network analysis showed 4 (miR-760, miR-3175, miR-885-3p, and miR-766-3p) of the 14 EV-miRNAs formed a regulatory "hub", suggesting co-targeting of specific gene pathways. In silico prediction identified a number of pathways important in lung injury. Two miRNAs involved in regulation of the cystic fibrosis transmembrane conductance regulator (CFTR), miRNA-145-5p and miRNA-138-5p, were also significantly increased in ARDS BALF-exposed hMSCs EVs. Functionally, EVs from hMSCs exposed to either ARDS or HV BALF had differential effects on CFTR Cl- secretion by cultured primary human bronchial epithelial cells, an effect predicted to reduce mucociliary clearance. The potential clinical impact of these finding highlights the need for further studies assessing the role of hMSC-EV miRNAs in regulating lung inflammation and mucociliary clearance.
Collapse
Affiliation(s)
- Sara Rolandsson Enes
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA; Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.
| | - Irakli Dzneladze
- Interdepartmental Division of Critical Care, Department of Medicine and the Keenan Center for Biomedical Research, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Thomas H Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Samuel L Neff
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Lori Asarian
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Jayita Barua
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Tobias Tertel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen Germany
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen Germany
| | - Nicolas Pereyra
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Canada, The University of British Columbia Centre for Blood Research, Vancouver, Canada
| | - David H McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, USA
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Canada
| | - Erica Acton
- Interdepartmental Division of Critical Care, Department of Medicine and the Keenan Center for Biomedical Research, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Alix Ashare
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA; Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Kathleen D Liu
- Departments of Medicine and Anesthesiology and the Cardiovascular Research Institute, University of California San Francisco
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queens University, Belfast, UK
| | - Karen English
- Cellular Immunology Laboratory, Biology Department, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Bruce A Stanton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Michael A Matthay
- Departments of Medicine and Anesthesiology and the Cardiovascular Research Institute, University of California San Francisco
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care, Department of Medicine and the Keenan Center for Biomedical Research, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Daniel J Weiss
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
4
|
Hoseini SM, Montazeri F. The influence of cell source on the senescence of human mesenchymal stem/stromal cells. Hum Cell 2025; 38:87. [PMID: 40221541 DOI: 10.1007/s13577-025-01213-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
While mesenchymal stem/stromal cells (MSCs) exhibit the ability to self-renew, they are not immortal; they eventually reach a point of irreversible growth cessation and functional deterioration following a limited series of population doublings, referred to as replicative senescence. When evaluated according to the criteria set by the International Society for Cell Therapy (ISCT), MSCs show significant differences in their senescence patterns and other characteristics related to their phenotype and function. These differences are attributed to the source of the MSCs and the conditions in which they are grown. MSCs derived from fetal or adult sources have variations in their genome stability, as well as in the expression and epigenetic profile of the cells, which in turn affects their secretome. Understanding the key factors of MSC senescence based on cell source can help to develop effective strategies for regulating senescence and improving the therapeutic potential.
Collapse
Affiliation(s)
- Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, No. 1. Safaeyeh. Bou-Al Ave., Yazd, 8916877391, Iran.
| |
Collapse
|
5
|
Hoseini SM, Montazeri F. Cell origin and microenvironment: The players of differentiation capacity in human mesenchymal stem cells. Tissue Cell 2025; 93:102709. [PMID: 39765135 DOI: 10.1016/j.tice.2024.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 03/05/2025]
Abstract
Mesenchymal stem cells (MSCs) have several important properties that make them desirable for regenerative medicine. These properties include immunomodulatory ability, growth factor production, and differentiation into various cell types. Despite extensive research and promising results in clinical trials, our understanding of MSC biology, their mechanism of action, and their targeted and routine use in clinics is limited. Differentiation of human MSCs (hMSCs) is a complex process influenced by various elements such as growth factors, pharmaceutical compounds, microRNAs, 3D scaffolds, and mechanical and electrical stimulation. Research has shown that different culture conditions can affect the differentiation potential of hMSCs obtained from multiple fetal and adult sources. Additionally, it seems that what affects the differentiation capacities of these cells is their secretory characteristics, which are influenced by the origin of the cells and the local microenvironment where the cells are located. The review can provide insights into the microenvironment-based mechanisms involved in MSC differentiation, which can be valuable for future therapeutic applications.
Collapse
Affiliation(s)
- Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| |
Collapse
|
6
|
Cui C, Lin F, Xia L, Zhang X. Mesenchymal stem cells therapy for the treatment of non-union fractures: a systematic review and meta-analysis. BMC Musculoskelet Disord 2025; 26:245. [PMID: 40069694 PMCID: PMC11900535 DOI: 10.1186/s12891-025-08365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND This meta-analysis aimed to pool the existing evidence to determine the clinical efficacy and safety of mesenchymal stem cells (MSC) in patients with non-unions. METHODS A systematic search in PubMed and Scopus was performed until October 2024 to gather pertinent studies. The inclusion criteria included participants with non-unions, the intervention of MSC administration, a comparator of standard treatment (bone graft), and outcomes focused on healing rate, healing time, or side effects. The Jadad score Newcastle-Ottawa Scale (NOS) was used to assess the risk of bias in randomized and non-randomized studies, respectively. Moreover, GRADE criteria were used to assess the quality of evidence. Using a random effects model, odds ratios (OR) with 95% confidence intervals (CIs) were calculated for healing and complication rates, while standardized mean differences (SMD) with their 95% CIs were used to assess the impact of MSC therapy on bone union time. RESULTS Twenty-one studies, with 866 patients, were included. The bone healing rates were 44% at 3 months, 73% at 6 months, 90% at 9 months, and 86% at 12 months, eventually reaching 91% after 12 months of follow-up. MSC therapy, with or without scaffolds, was linked to higher odds of bone healing rate at 3 and 6 months, compared to bone grafts as the standard care (OR = 1.69). The time to union following the treatment was 6.30 months (95%CI: 86-96%), with patients treated with MSC/Scaffold experiencing a shorter time compared to MSC alone (5.85 vs. 6.36 months). MSC therapy significantly decreased bone union time (SMD:-0.54 months, 95% CI: -0.75 to -0.33). The complication rate was 1% (MSC/Scaffold: 0%, MSC alone: 2%), with MSC alone or MSC/Scaffold showing a lower risk than the standard care (OR = 0.41, 95% CI: 0.22-0.78). CONCLUSION MSC is a potential adjunct therapy for patients with non-union fractures. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Cunbao Cui
- Department of Joint Surgery, Central Hospital Affiliated to Shandong First Medical University, NO. 105, Jiefang Road, Jinan, 250013, China
| | - Feng Lin
- Department of Joint Surgery, Central Hospital Affiliated to Shandong First Medical University, NO. 105, Jiefang Road, Jinan, 250013, China
| | - Liang Xia
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, NO. 105, Jiefang Road, Jinan, 250013, China
| | - Xinguang Zhang
- Department of Joint Surgery, Central Hospital Affiliated to Shandong First Medical University, NO. 105, Jiefang Road, Jinan, 250013, China.
| |
Collapse
|
7
|
Akbarzadeh A, Gerami MH, Farrokhi MR, Shapoori S, Jafarinia M. Therapeutic prospects of microRNAs derived from mesenchymal stem cell extracellular vesicles in rheumatoid arthritis: a comprehensive overview. Mol Cell Biochem 2025; 480:1275-1286. [PMID: 39105963 DOI: 10.1007/s11010-024-05082-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by inflammatory joint damage. Recent studies have focused on the significance of microRNAs (miRNAs) in the pathogenesis of RA. Mesenchymal stem cells (MSCs) have emerged as a potential therapeutic option for RA based on their regenerative and immunomodulatory properties. MSCs release extracellular vesicles (EVs) containing miRNAs that can modulate immune and inflammatory responses. This article provides a comprehensive overview of the current evidence on the existence of various MSCs-derived miRNAs involved in the pathophysiology, characterization, and treatment of RA. An overview of the miRNA profiles in MSC-EVs is provided, along with an examination of their impact on various cell types implicated in RA pathogenesis, including synovial fibroblasts, macrophages, and T cells. Furthermore, the therapeutic capability of MSC-EVs for miRNA-based therapies in RA is discussed. In total, this review can present an extensive view of the complex interaction between EVs and MSC-derived miRNAs in RA and thus suggest valuable strategies for developing new therapeutic approaches to target this debilitating disease.
Collapse
Affiliation(s)
- Armin Akbarzadeh
- Department of Orthopedic Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Gerami
- Department of Orthopedic Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Reza Farrokhi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shima Shapoori
- Center for Research in Medical Devices (CÚRAM), University of Galway, Galway, Ireland
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Nada AH, Ibrahim IA, Oteri V, Shalabi L, Asar NK, Aqeilan SR, Hafez W. Safety and efficacy of umbilical cord mesenchymal stem cells in the treatment of type 1 and type 2 diabetes mellitus: a systematic review and meta-analysis. Expert Rev Endocrinol Metab 2025; 20:107-117. [PMID: 39905688 DOI: 10.1080/17446651.2025.2457474] [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: 08/03/2024] [Accepted: 11/22/2024] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Many patients struggle to control glucose without side effects. Due to their immunomodulatory and regenerative properties, mesenchymal stem cells (MSCs) might treat Diabetes Mellitus (DM). The authors employed this meta-analysis to evaluate the efficacy and safety of umbilical cord MSCs (UCMSCs) for DM management. METHODS The PubMed, Cochrane, WOS, Embase, and Scopus databases were searched for randomized controlled trials (RCTs) investigating the effects of UCMSCs on DM (Types 1, 2) till January 2024. Patient demographics, interventions, and outcomes, including glycated hemoglobin (HbA1c%), C-peptide levels, and insulin requirements, were extracted. A comprehensive meta-analysis software was used. RESULTS Eight CTs of 334 patients (172 experimental and 162 controls) were included. UMSCs treatment substantially lowered HbA1c levels (MD = -1.06, 95% CI [-1.27, -0.85], p < 0.00001) with consistent outcomes (i2 = 0%, p = 0.43). Fasting C-peptide levels were heterogeneous but favored placebo (MD = 0.35, 95% CI [0.15, 0.56], p = 0.0007). In T1D patients, daily insulin requirements decreased considerably (MD = -0.24, 95% CI [-0.29, -0.18], p < 0.00001), with heterogeneity addressed by sensitivity analysis. CONCLUSION UMSCs therapy reduced HbA1c and insulin requirements, and increased C-peptide levels. Multicenter clinical trials are required to confirm the long-term efficacy and safety of UMSC therapy.
Collapse
Affiliation(s)
| | - Ismail A Ibrahim
- Faculty of Health Sciences, Fenerbahce University, Istanbul, Turkey
| | - Vittorio Oteri
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, Catania, Italy
| | - Laila Shalabi
- Faculty of Medicine, Gharyan University, Gharyan, Libya
| | | | | | - Wael Hafez
- Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
- NMC Royal Hospital, Khalifa City, Abu Dhabi, UAE
| |
Collapse
|
9
|
Patel AA, Shafie A, Mohamed AH, Ali SAJ, Tayeb FJ, Waggiallah HA, Ahmad I, Sheweita SA, Muzammil K, AlShahrani AM, Al Abdulmonem W. The promise of mesenchymal stromal/stem cells in erectile dysfunction treatment: a review of current insights and future directions. Stem Cell Res Ther 2025; 16:98. [PMID: 40012076 PMCID: PMC11866689 DOI: 10.1186/s13287-025-04221-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
Erectile dysfunction is a common and multifactorial condition that significantly impacts men's quality of life. Traditional treatments, such as phosphodiesterase type 5 inhibitors (PDE5i), often fail to provide lasting benefits, particularly in patients with underlying health conditions. In recent years, regenerative medicine, particularly stem cell therapies, has emerged as a promising alternative for managing erectile dysfunction. This review explores the potential of mesenchymal stromal/stem cells (MSCs) and their paracrine effects, including extracellular vesicles (EVs), in the treatment of erectile dysfunction. MSCs have shown remarkable potential in promoting tissue repair, reducing inflammation, and regenerating smooth muscle cells, offering therapeutic benefits in models of erectile dysfunction. Clinical trials have demonstrated positive outcomes in improving erectile function and other clinical parameters. This review highlights the promise of MSC therapy for erectile dysfunction, discusses existing challenges, and emphasizes the need for continued research to refine these therapies and improve long-term patient outcomes.
Collapse
Affiliation(s)
- Ayyub Ali Patel
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | | | - Faris J Tayeb
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkarj, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Salah Ahmed Sheweita
- Department of Clinical Biochemistry, Faculty of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait, King Khalid University, 62561, Abha, Saudi Arabia
| | - Abdullah M AlShahrani
- Department of Basic Medical Science, College of Applied Medical Sciences, Khamis Mushait, King Khalid University (KKU), 62561, Abha, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| |
Collapse
|
10
|
Tian Y, Jin M, Ye N, Gao Z, Jiang Y, Yan S. Mesenchymal stem cells-derived exosomes attenuate mouse non-heart-beating liver transplantation through Mir-17-5p-regulated Kupffer cell pyroptosis. Stem Cell Res Ther 2025; 16:57. [PMID: 39920844 PMCID: PMC11806715 DOI: 10.1186/s13287-025-04169-w] [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: 07/23/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Liver transplantation is the most effective treatment for end-stage liver disease. However, the shortage of donor livers has become a significant obstacle to the advancement of liver transplantation. Mesenchymal stem cells-derived exosomes (MSCs-Exo) have been extensively investigated in liver diseases. However, the underlying mechanisms of how they can protect organ donation after cardiac death (DCD) livers remain unclear. METHODS In this study, an arterialized mouse non-heart-beating (NHB) liver transplantation model was used to investigate the effect of MSCs-Exo on NHB liver transplantation. The survival rates, histology, pro-inflammatory cytokine and chemokine expression, and underlying mechanisms were investigated. RESULTS The infusion of MSCs-Exo reduced the injury to DCD liver graft tissue. In vitro and in vivo experiments demonstrated that MSCs-Exo could inhibit hydrogen peroxide-induced pyroptosis of Kupffer cells. We found that miR-17-5p was significantly abundant in MSCs-Exo, targeting and regulating the TXNIP expression. This action inhibited NLRP3-mediated pyroptosis of Kupffer cells through the classical Caspase1-dependent pathway, alleviating DCD liver graft injury. CONCLUSION Our study elucidated a protective role for MSCs-Exo in a NHB liver transplantation model. This mechanism provides a theoretical basis and new strategies for the clinical application of MSCs-Exo to improve liver graft quality and alleviate the organ shortage in liver transplantation.
Collapse
Affiliation(s)
- Yang Tian
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Jie-Fang Road #88, Hangzhou, Zhejiang Province, 310009, China
| | - Ming Jin
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Jie-Fang Road #88, Hangzhou, Zhejiang Province, 310009, China
| | - Nanwei Ye
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Zhenzhen Gao
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Jie-Fang Road #88, Hangzhou, Zhejiang Province, 310009, China
| | - Yuancong Jiang
- Department of Surgery, Shaoxing People's Hospital, Zhejiang University School of Medicine, Zhong-Xing North Road #568, Shaoxing, Zhejiang Province, 312000, China.
| | - Sheng Yan
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Jie-Fang Road #88, Hangzhou, Zhejiang Province, 310009, China.
| |
Collapse
|
11
|
Kim OH, Kang H, Chang ES, Lim Y, Seo YJ, Lee HJ. Extended protective effects of three dimensional cultured human mesenchymal stromal cells in a neuroinflammation model. World J Stem Cells 2025; 17:101485. [PMID: 39866897 PMCID: PMC11752454 DOI: 10.4252/wjsc.v17.i1.101485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/11/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Human mesenchymal stromal cells (MSCs) possess regenerative potential due to pluripotency and paracrine functions. However, their stemness and immunomodulatory capabilities are sub-optimal in conventional two-dimensional (2D) culture. AIM To enhance the efficiency and therapeutic efficacy of MSCs, an in vivo-like 3D culture condition was applied. METHODS MSCs were cultured on polystyrene (2D) or in a gellan gum-based 3D system. In vitro, prostaglandin-endoperoxide synthase 2, indoleamine-2,3-dioxygenase, heme oxygenase 1, and prostaglandin E synthase gene expression was quantified by quantitative real-time polymerase chain reaction. MSCs were incubated with lipopolysaccharide (LPS)-treated mouse splenocytes, and prostaglandin E2 and tumor necrosis factor-alpha levels were measured by enzyme linked immunosorbent assay. In vivo, LPS was injected into the lateral ventricle of mouse brain, and MSCs were administered intravenously the next day. Animals were sacrificed and analyzed on days 2 and 6. RESULTS Gellan gum polymer-based 3D culture significantly increased expression of octamer-binding transcription factor 4 and Nanog homeobox stemness markers in human MSCs compared to 2D culture. This 3D environment also heightened expression of cyclooxygenase-2 and heme-oxygenase 1, enzymes known for immunomodulatory functions, including production of prostaglandins and heme degradation, respectively. MSCs in 3D culture secreted more prostaglandin E2 and effectively suppressed tumor necrosis factor-alpha release from LPS-stimulated splenocytes and surpassed the efficiency of MSCs cultured in 2D. In a murine neuroinflammation model, intravenous injection of 3D-cultured MSCs significantly reduced ionized calcium-binding adaptor molecule 1 and glial fibrillary acidic protein expression, mitigating chronic inflammation more effectively than 2D-cultured MSCs. CONCLUSION The microenvironment established in 3D culture serves as an in vivo mimetic, enhancing the immunomodulatory function of MSCs. This suggests that engineered MSCs hold significant promise a potent tool for cell therapy.
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul 06974, South Korea
| | - Hana Kang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, South Korea
| | - Eun Seo Chang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, South Korea
| | - Younghyun Lim
- Department of Life Science, Chung-Ang University, Seoul 06974, South Korea
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Seoul 06974, South Korea
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul 06974, South Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul 06974, South Korea.
| |
Collapse
|
12
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
13
|
Serack FE, Fennell KA, Iliopoulos C, Walker JT, Ronald JA, Amsden BG, Hess DA, Flynn LE. Probing the effects of polysaccharide hydrogel composition on the viability and pro-angiogenic function of human adipose-derived stromal cells. J Biomed Mater Res A 2025; 113:e37800. [PMID: 39304971 DOI: 10.1002/jbm.a.37800] [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: 05/12/2024] [Revised: 08/02/2024] [Accepted: 09/11/2024] [Indexed: 12/26/2024]
Abstract
Cell therapies harnessing the pro-vascular regenerative capacities of mesenchymal stromal cell (MSC) populations, including human adipose-derived stromal cells (hASCs), have generated considerable interest as an emerging treatment strategy for peripheral arterial disease (PAD) and its progression to critical limb ischemia (CLI). There is evidence to support that polysaccharide hydrogels can enhance therapeutic efficacy when applied as minimally-invasive delivery systems to support MSC survival and retention within ischemic tissues. However, there has been limited research to date on the effects of hydrogel composition on the phenotype and function of encapsulated cell populations. Recognizing this knowledge gap, this study compared the pro-angiogenic function of hASCs encapsulated in distinct but similarly-modified natural polysaccharide hydrogels composed of methacrylated glycol chitosan (MGC) and methacrylated hyaluronic acid (MHA). Initial in vitro studies confirmed high viability (>85%) of the hASCs following encapsulation and culture in the MGC and MHA hydrogels over 14 days, with a decrease in the cell density observed over time. Moreover, higher levels of a variety of secreted pro-angiogenic and immunomodulatory factors were detected in conditioned media samples collected from the hASCs encapsulated in the MGC-based hydrogels compared to the MHA hydrogels. Subsequent testing focused on comparing hASC delivery within the MGC and MHA hydrogels to saline controls in a femoral artery ligation-induced CLI (FAL-CLI) model in athymic nu/nu mice over 28 days. For the in vivo studies, the hASCs were engineered to express tdTomato and firefly luciferase to quantitatively compare the efficacy of the two platforms in supporting the localized retention of viable hASCs through longitudinal cell tracking with bioluminescence imaging (BLI). Interestingly, hASC retention was significantly enhanced when the cells were delivered in the MHA hydrogels as compared to the MGC hydrogels or saline. However, laser Doppler perfusion imaging (LDPI) indicated that the restoration of hindlimb perfusion was similar between the treatment groups and controls. These findings were corroborated by endpoint immunofluorescence (IF) staining showing similar levels of CD31+ cells in the ligated limbs at 28 days in all groups. Overall, this study demonstrates that enhanced MSC retention may be insufficient to augment vascular regeneration, emphasizing the complexity of designing biomaterials platforms for MSC delivery for therapeutic angiogenesis. In addition, the data points to a potential challenge in approaches that seek to harness the paracrine functionality of MSCs, as strategies that increase the secretion of immunomodulatory factors that can aid in regeneration may also lead to more rapid MSC clearance in vivo.
Collapse
Affiliation(s)
- Fiona E Serack
- School of Biomedical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Kaylee A Fennell
- Department of Chemical & Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Christina Iliopoulos
- Department of Chemical & Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario, Canada
| | - John T Walker
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - John A Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | - Brian G Amsden
- Department of Chemical Engineering, Faculty of Engineering, Queen's University, Kingston, Ontario, Canada
| | - David A Hess
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Lauren E Flynn
- School of Biomedical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario, Canada
- Department of Chemical & Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
14
|
Garmany A, Arrell DK, Yamada S, Jeon R, Behfar A, Park S, Terzic A. Decoded cardiopoietic cell secretome linkage to heart repair biosignature. Stem Cells Transl Med 2024; 13:1144-1159. [PMID: 39259666 PMCID: PMC11555478 DOI: 10.1093/stcltm/szae067] [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: 04/09/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
Cardiopoiesis-primed human stem cells exert sustained benefit in treating heart failure despite limited retention following myocardial delivery. To assess potential paracrine contribution, the secretome of cardiopoiesis conditioned versus naïve human mesenchymal stromal cells was decoded by directed proteomics augmented with machine learning and systems interrogation. Cardiopoiesis doubled cellular protein output generating a distinct secretome that segregated the conditioned state. Altering the expression of 1035 secreted proteins, cardiopoiesis reshaped the secretome across functional classes. The resolved differential cardiopoietic secretome was enriched in mesoderm development and cardiac progenitor signaling processes, yielding a cardiovasculogenic profile bolstered by upregulated cardiogenic proteins. In tandem, cardiopoiesis enhanced the secretion of immunomodulatory proteins associated with cytokine signaling, leukocyte migration, and chemotaxis. Network analysis integrated the differential secretome within an interactome of 1745 molecules featuring prioritized regenerative processes. Secretome contribution to the repair signature of cardiopoietic cell-treated infarcted hearts was assessed in a murine coronary ligation model. Intramyocardial delivery of cardiopoietic cells improved the performance of failing hearts, with undirected proteomics revealing 50 myocardial proteins responsive to cell therapy. Pathway analysis linked the secretome to cardiac proteome remodeling, pinpointing 17 cardiopoiesis-upregulated secretome proteins directly upstream of 44% of the cell therapy-responsive cardiac proteome. Knockout, in silico, of this 22-protein secretome-dependent myocardial ensemble eliminated indices of the repair signature. Accordingly, in vivo, cell therapy rendered the secretome-dependent myocardial proteome of an infarcted heart indiscernible from healthy counterparts. Thus, the secretagogue effect of cardiopoiesis transforms the human stem cell secretome, endows regenerative competency, and upregulates candidate paracrine effectors of cell therapy-mediated molecular restitution.
Collapse
Affiliation(s)
- Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - D Kent Arrell
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Section of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Sungjo Park
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
15
|
Jaffet J, Singh V, Schrader S, Mertsch S. The Potential Role of Exosomes in Ocular Surface and Lacrimal Gland Regeneration. Curr Eye Res 2024:1-14. [PMID: 39508276 DOI: 10.1080/02713683.2024.2424265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/01/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE Dry eye disease (DED), a multifactorial disease of the lacrimal system, manifests itself in patients with various symptoms such as itching, inflammation, discomfort and visual impairment. In its most severe forms, it results in the breakdown of the vital tissues of lacrimal functional unit and carries the risk of vision loss. Despite the frequency of occurrence of the disease, there are no effective curative treatment options available to date. Treatment using stem cells and its secreted factors could be a promising approach in the regeneration of damaged tissues of ocular surface. The treatment using secreted factors as well as extracellular vesicles has been demonstrated beneficial effects in various ocular surface diseases. This review provides insights on the usage of stem cell derived exosomes as a promising therapy against LG dysfunction induced ADDE for ocular surface repair. METHODS In order to gain an overview of the existing research in this field, literature search was carried out using the PubMed, Medline, Scopus and Web of Science databases. This review is based on 164 publications until June 2024 and the literature search was carried out using the key words "exosomes", "lacrimal gland regeneration", "exosomes in lacrimal dysfunction". RESULTS The literature and studies till date suggest that exosomes and other secreted factors from stem cells have demonstrated beneficial effects on damaged ocular tissues in various ocular surface diseases. Exosomal cargo plays a crucial role in regenerating tissues by promoting homeostasis in the lacrimal system, which is often compromised in severe cases of dry eye disease. Exosome therapy shows promise as a regenerative therapy, potentially addressing the lack of effective curative treatments available for patients with dry eye disease. CONCLUSION Stem cell-derived exosomes represent a promising, innovative approach as a new treatment option for ADDE. By targeting lacrimal gland dysfunction and enhancing ocular surface repair, exosome therapy offers potential for significant advances in dry eye disease management. Future research is needed to refine the application of this therapy, optimize delivery methods, and fully understand its long-term efficacy in restoring ocular health.
Collapse
Affiliation(s)
- Jilu Jaffet
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
- LV Prasad Eye Institute, Centre for Ocular Regeneration, Hyderabad, Telangana, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Vivek Singh
- LV Prasad Eye Institute, Centre for Ocular Regeneration, Hyderabad, Telangana, India
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
| |
Collapse
|
16
|
Ding K, Kong J, Li L, Selaru FM, Parian A, Mao HQ. Current and emerging therapeutic strategies for perianal fistula in Crohn's disease patients. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:159-182. [PMID: 39521599 PMCID: PMC11753511 DOI: 10.1016/bs.apha.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The long-term remission rates achieved with current treatment options for Crohn's disease with perianal fistula (CD-PAF)-including antibiotics, biologics, immunomodulators, and Janus kinase inhibitors, often combined with advanced surgical interventions-remain unsatisfactory. This chapter explores several innovative biomaterials-based solutions, such as plugs, adhesives, fillers, and stem cell-based therapies. The key approaches and treatment outcomes of these advanced therapies are examined, focusing on their ability to modulate the immune response, promote tissue healing, and improve patient outcomes. Additionally, the chapter discusses future directions, including the optimization of biomaterial designs, enhancement of delivery and retention of regenerative therapies, and a deeper understanding of the underlying mechanisms of healing.
Collapse
Affiliation(s)
- Kailei Ding
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jiayuan Kong
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ling Li
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Florin M Selaru
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa Parian
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
17
|
Xu S, Zhang M, Wang R, Zhang J, Wang C, Xie L, Zhao W. Spatial dimension cues derived from fibrous scaffolds trigger mechanical activation to potentiate the paracrine and regenerative functions of MSCs via the FAK-PI3K/AKT axis. Acta Biomater 2024:S1742-7061(24)00631-7. [PMID: 39461692 DOI: 10.1016/j.actbio.2024.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/29/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Secretomes from mesenchymal stem cells (MSCs) have significant therapeutic potential and could be the basis for future MSCs treatments. Innovative design of the topology of biomaterials, which mechanically regulate cell behavior and function, can tremendously improve the efficacy of stem cell therapy. However, how spatial dimension cues derived from specific topology command cell mechanotransduction to regulate the paracrine function of MSCs remains unknown. In this study, the three-dimensional (3D) fibrous constructs with box-like pores and precise strand spacing from 150 µm down to only 40 µm were manufactured using melt electrowriting (MEW), which were used to systematically investigate the spatial dimension cues-triggered mechanotransduction of adipose-derived mesenchymal stem cells (Ad-MSCs) and their impact on the paracrine and regeneration function of Ad-MSCs. The results demonstrated that spatial instructions from the 3D fibrous constructs could influence the spatial reorganization of the cytoskeleton, resulting in cell elongation and augmented immunomodulatory and angiogenic paracrine effects of Ad-MSCs, which was most pronounced at a minimum strand spacing of 40 µm. Besides, mechanical activation of the FAK-PI3K/AKT axis significantly enhanced the paracrine function of Ad-MSCs. In vivo experiments demonstrated that the Ad-MSCs trained using well-defined 3D fibrous constructs with a strand spacing of 40 µm significantly promoted skin regeneration via paracrine signals. In conclusion, this study provides a new horizon for deciphering space dimension insights into the interactional mechanisms of mechanotransduction in regulating cell function, which has inspired innovations in biomaterials for improving tissue regeneration. STATEMENT OF SIGNIFICANCE: This study emphasized that designing cell-scale spatial dimension cues to command mechanical activation via the FAK-PI3K/AKT axis could significantly enhance the paracrine and regenerative functions of Ad-MSCs. Paracrine signals of Ad-MSCs triggered by mechanical activation promoted skin repair and regeneration via the immunomodulation and angiogenesis. The proposed mechanobiological signal transduction triggered by spatial dimensional cues, which potentiates the paracrine and regenerative functions of Ad-MSCs, is a promising engineering strategy and is expected to provide new inspirations for the development of biomaterials based on biophysical signals for cellular behavior modulation.
Collapse
Affiliation(s)
- Shixin Xu
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Miaomiao Zhang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Ruoying Wang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jinxin Zhang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chengwei Wang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Li Xie
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Wen Zhao
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
18
|
Areny-Balagueró A, Camprubí-Rimblas M, Campaña-Duel E, Solé-Porta A, Ceccato A, Roig A, Laffey JG, Closa D, Artigas A. Priming Mesenchymal Stem Cells with Lipopolysaccharide Boosts the Immunomodulatory and Regenerative Activity of Secreted Extracellular Vesicles. Pharmaceutics 2024; 16:1316. [PMID: 39458645 PMCID: PMC11510928 DOI: 10.3390/pharmaceutics16101316] [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: 09/05/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Mesenchymal stem cells (MSCs)-derived extracellular vesicles (EVs) have been proposed as an alternative to live-cell administration for Acute Respiratory Distress Syndrome (ARDS). MSC-EVs can be chiefly influenced by the environment to which the MSCs are exposed. Here, lipopolysaccharide (LPS) priming of MSCs was used as a strategy to boost the natural therapeutic potential of the EVs in acute lung injury (ALI). Methods: The regenerative and immunemodulatory effect of LPS-primed MSC-EVs (LPS-EVs) and non-primed MSC-EVs (C-EVs) were evaluated in vitro on alveolar epithelial cells and macrophage-like THP-1 cells. In vivo, ALI was induced in adult male rats by the intrapulmonary instillation of HCl and LPS. Rats (n = 8 to 22/group) were randomized to receive a single bolus (1 × 108 particles) of LPS-EVs, C-EVs, or saline. Lung injury severity was assessed at 72 h in lung tissue and bronchoalveolar lavage. Results: In vitro, LPS-EVs improved wound regeneration and attenuated the inflammatory response triggered by the P. aeruginosa infection, enhancing the M2 macrophage phenotype. In in vivo studies, LPS-EVs, but not C-EVs, significantly decreased the neutrophilic infiltration and myeloperoxidase (MPO) activity in lung tissue. Alveolar macrophages from LPS-EVs-treated animals exhibited a reduced expression of CXCL-1, a key neutrophil chemoattractant. However, both C-EVs and LPS-EVs reduced alveolar epithelial and endothelial permeability, mitigating lung damage. Conclusions: EVs from LPS-primed MSCs resulted in a better resolution of ALI, achieving a greater balance in neutrophil infiltration and activation, while avoiding the complete disruption of the alveolar barrier. This opens new avenues, paving the way for the clinical implementation of cell-based therapies.
Collapse
Affiliation(s)
- Aina Areny-Balagueró
- Critical Care Research Center, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.C.); (A.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Camprubí-Rimblas
- Critical Care Research Center, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.C.); (A.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena Campaña-Duel
- Critical Care Research Center, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.C.); (A.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Anna Solé-Porta
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; (A.S.-P.); (A.R.)
| | - Adrián Ceccato
- Critical Care Research Center, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.C.); (A.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Intensive Care Unit, Hospital Universitari Sagrat Cor, Grupo Quironsalud, 08029 Barcelona, Spain
| | - Anna Roig
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain; (A.S.-P.); (A.R.)
| | - John G. Laffey
- REMEDI, CÚRAM Centre for Medical Device Research, University of Galway, H91 TK33 Galway, Ireland;
| | - Daniel Closa
- Institut d’Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain;
| | - Antonio Artigas
- Critical Care Research Center, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain; (M.C.-R.); (E.C.-D.); (A.C.); (A.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servei de Medicina Intensiva, Corporació Sanitària i Universitària Parc Taulí, 08208 Sabadell, Spain
| |
Collapse
|
19
|
Türküner MS, Yazıcı A, Özcan F. SIK2 Controls the Homeostatic Character of the POMC Secretome Acutely in Response to Pharmacological ER Stress Induction. Cells 2024; 13:1565. [PMID: 39329749 PMCID: PMC11430698 DOI: 10.3390/cells13181565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The neuronal etiology of obesity is centered around a diet-induced inflammatory state in the arcuate nucleus of the hypothalamus, which impairs the functionality of pro-opiomelanocortine neurons (POMCs) responsible for whole-body energy homeostasis and feeding behavior. Intriguingly, systemic salt inducible kinase 2 (SIK2) knockout mice demonstrated reduced food intake and energy expenditure along with modestly dysregulated metabolic parameters, suggesting a causal link between the absence of SIK2 activity in POMCs and the observed phenotype. To test this hypothesis, we conducted a comparative secretomics study from POMC neurons following pharmacologically induced endoplasmic reticulum (ER) stress induction, a hallmark of metabolic inflammation and POMC dysregulation in diet-induced obese (DIO) mice. Our data provide significant in vitro evidence for the POMC-specific SIK2 activity in controlling energy metabolism and feeding in DIO mice by regulating the nature of the related POMC secretome. Our data also suggest that under physiological stress conditions, SIK2 may act as a gatekeeper for the secreted inflammatory factors and signaling molecules critical for cellular survival and energy homeostasis. On the other hand, in the absence of SIK2, the gate opens, leading to a surge of inflammatory cytokines and apoptotic cues concomitant with the dysregulation of POMC neurons.
Collapse
Affiliation(s)
- Mehmet Soner Türküner
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| | - Ayşe Yazıcı
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| | - Ferruh Özcan
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| |
Collapse
|
20
|
Weiss DJ. Unraveling the Complexities of Mesenchymal Stromal Cell-based Therapies: One Size Doesn't Fit All. Am J Respir Crit Care Med 2024; 210:709-711. [PMID: 38843143 PMCID: PMC11418884 DOI: 10.1164/rccm.202405-0961ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024] Open
Affiliation(s)
- Daniel J Weiss
- Department of Medicine
- Department of Bioengineering University of Vermont Burlington, Vermont
| |
Collapse
|
21
|
Chen B, Zhu X, Zhang D, Zhu Z, Ye Q, Guo J. Adipose-derived mesenchymal stem cells suppress fibroblast proliferation of hypertrophic scar through CCL5 and CXCL12. Arch Dermatol Res 2024; 316:527. [PMID: 39153095 DOI: 10.1007/s00403-024-03289-2] [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: 07/08/2024] [Revised: 07/08/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND AND OBJECTIVE Adipose-derived mesenchymal stem cells (ADSCs) can accelerate wound healing, reduce scar formation, and inhibit hypertrophic scar (HTS). ADSCs can secrete a large amount of CCL5, and CCL5 has been proved to be pro-inflammatory and pro-fibrotic. CXCL12 (SDF-1) is a key chemokine that promotes stem cell migration and survival. Therefore, this study selected normal skin and HTS conditioned medium to simulate different microenvironments, and analyzed the effects of different microenvironments on the expression of CCL5 and CXCL12 in human ADSCs (hADSCs). MATERIALS AND METHODS hADSCs with silenced expression of CCL5 and CXCL12 were co-cultured with hypertrophic scar fibroblasts to verify the effects of CCL5 and CXCL12 in hADSCs on the proliferation ability of hypertrophic scar fibroblasts. A mouse model of hypertrophic scar was established to further confirm the effect of CCL5 and CXCL12 in hADSCs on hypertrophic scar formation. RESULTS CCL5 level was found to be significantly high in hADSCs cultured in HTS conditioned medium. CXCL12 in HTS group was prominently lowly expressed compared with the normal group. Inhibition of CCL5 in hADSCs enhanced the effects of untreated hADSCs on proliferation of HTS fibroblasts while CXCL12 knockdown exerted the opposite function. Inhibition of CCL5 in hADSCs increased the percentage of HTS fibroblasts in the G0/G1 phase while down-regulation of CXCL12 decreased those. Meanwhile, the down-regulated levels of fibroblast markers including collagen I, collagen III, and α-SMA induced by CCL5 knockdown were significantly up-regulated by CXCL12 inhibition. hADSCs alleviate the HTS of mice through CCL5 and CXCL12. CONCLUSION In summary, our results demonstrated that hADSCs efficiently cured HTS by suppressing proliferation of HTS fibroblasts, which may be related to the inhibition of CXCL12 and elevation of CCL5 in hADSCs, suggesting that hADSCs may provide an alternative therapeutic approach for the treatment of HTS.
Collapse
Affiliation(s)
- Bo Chen
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Xiongxiang Zhu
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Dongmei Zhang
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Zhensen Zhu
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China
| | - Qian Ye
- Department of Orthopaedics, Shenzhen Hospital of Southern Medical University, Shenzhen, 518000, Guangdong, China
| | - Jingdong Guo
- Department of Burns and Plastic Surgery, Shenzhen Hospital of Southern Medical University, No.1333, Xinhu Road, Baoan District, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
22
|
Chen Y, Zhang Z, Li Z, Wu W, Lan S, Yan T, Mei K, Qiao Z, Wang C, Bai C, Li Z, Wu S, Wang J, Zhang Q. Dynamic nanomechanical characterization of cells in exosome therapy. MICROSYSTEMS & NANOENGINEERING 2024; 10:97. [PMID: 39015940 PMCID: PMC11251037 DOI: 10.1038/s41378-024-00735-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/20/2024] [Accepted: 05/23/2024] [Indexed: 07/18/2024]
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) have been confirmed to enhance cell proliferation and improve tissue repair. Exosomes release their contents into the cytoplasmic solution of the recipient cell to mediate cell expression, which is the main pathway through which exosomes exert therapeutic effects. The corresponding process of exosome internalization mainly occurs in the early stage of treatment. However, the therapeutic effect of exosomes in the early stage remains to be further studied. We report that the three-dimensional cell traction force can intuitively reflect the ability of exosomes to enhance the cytoskeleton and cell contractility of recipient cells, serving as an effective method to characterize the therapeutic effect of exosomes. Compared with traditional biochemical methods, we can visualize the early therapeutic effect of exosomes in real time without damage by quantifying the cell traction force. Through quantitative analysis of traction forces, we found that endometrial stromal cells exhibit short-term cell roundness accompanied by greater traction force during the early stage of exosome therapy. Further experiments revealed that exosomes enhance the traction force and cytoskeleton by regulating the Rac1/RhoA signaling pathway, thereby promoting cell proliferation. This work provides an effective method for rapidly quantifying the therapeutic effects of exosomes and studying the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ye Chen
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Zihan Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Ziwei Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Wenjie Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Shihai Lan
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Tianhao Yan
- Department of Cell Biology and Genetics, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Kainan Mei
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Zihan Qiao
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Chen Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Chuanbiao Bai
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Ziyan Li
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Shangquan Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Science, 15 Beisihuan West Road, Beijing, 100190 China
| | - Jianye Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Qingchuan Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| |
Collapse
|
23
|
Lee NK, Chang JW. Manufacturing Cell and Gene Therapies: Challenges in Clinical Translation. Ann Lab Med 2024; 44:314-323. [PMID: 38361427 PMCID: PMC10961620 DOI: 10.3343/alm.2023.0382] [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: 09/26/2023] [Revised: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
The safety and efficacy of both cell and gene therapies have been demonstrated in numerous preclinical and clinical trials. Chimeric antigen receptor T (CAR-T) cell therapy, which leverages the technologies of both cell and gene therapies, has also shown great promise for treating various cancers. Advancements in pertinent fields have also highlighted challenges faced while manufacturing cell and gene therapy products. Potential problems and obstacles must be addressed to ease the clinical translation of individual therapies. Literature reviews of representative cell-based, gene-based, and cell-based gene therapies with regard to their general manufacturing processes, the challenges faced during manufacturing, and QC specifications are limited. We review the general manufacturing processes of cell and gene therapies, including those involving mesenchymal stem cells, viral vectors, and CAR-T cells. The complexities associated with the manufacturing processes and subsequent QC/validation processes may present challenges that could impede the clinical progression of the products. This article addresses these potential challenges. Further, we discuss the use of the manufacturing model and its impact on cell and gene therapy.
Collapse
Affiliation(s)
- Na Kyung Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jong Wook Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
| |
Collapse
|
24
|
Darwish M, El Hajj R, Khayat L, Alaaeddine N. Stem Cell Secretions as a Potential Therapeutic Agent for Autism Spectrum Disorder: A Narrative Review. Stem Cell Rev Rep 2024; 20:1252-1272. [PMID: 38630359 DOI: 10.1007/s12015-024-10724-4] [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] [Accepted: 04/09/2024] [Indexed: 07/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental illness characterized by impaired social interaction and restricted repetitive behaviors or interests. The rising prevalence of ASD diagnosis has triggered a surge in research into investigating the underlying neuropathological processes and finding new therapeutic approaches. ASD is characterized by neuroinflammation and dysregulation of neuro-immune cross-talk, which suggests that stem cell treatment might be a potential therapeutic approach. The beneficial and restorative effects of stem cells are mainly due to their paracrine activity, in which stem cells generate and release extracellular vesicles such as exosomes and distinct secreted non-vesicle soluble proteins, including, growth factors, chemokines, cytokines, and immunomodulatory molecules referred to as the Secretome. In this paper, we reviewed the existing research exploring the therapeutic potential of stem cell secretome focusing on their role in addressing ASD pathology. Furthermore, we proposed a comprehensive mechanism of action for stem cell secretions, encompassing the broader secretome as well as the specific contribution of exosomes, in alleviating ASD neuropathology. Across the reviewed studies, exosomes and secreted soluble factors of the transplanted stem cell demonstrate a potential efficacy in ameliorating autistic-like behaviors. The proposed mechanism of action involves the modulation of signaling pathways implicated in neuroinflammation, angiogenesis, cellular apoptosis, and immunomodulation.
Collapse
Affiliation(s)
- Mariam Darwish
- Faculty of Medical Sciences, Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | | | | | - Nada Alaaeddine
- Dean of Health Sciences, Modern University for Business & Science, Beirut, Lebanon.
| |
Collapse
|
25
|
Sridharan D, Dougherty JA, Ahmed U, Sanghvi SK, Alvi SB, Park KH, Islam H, Knoblaugh SE, Singh H, Kirby ED, Khan M. Bioorthogonal non-canonical amino acid tagging to track transplanted human induced pluripotent stem cell-specific proteome. Stem Cell Res Ther 2024; 15:186. [PMID: 38926849 PMCID: PMC11210150 DOI: 10.1186/s13287-024-03792-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cells (hiPSCs) and their differentiated cell types have a great potential for tissue repair and regeneration. While the primary focus of using hiPSCs has historically been to regenerate damaged tissue, emerging studies have shown a more potent effect of hiPSC-derived paracrine factors on tissue regeneration. However, the precise contents of the transplanted hiPSC-derived cell secretome are ambiguous. This is mainly due to the lack of tools to distinguish cell-specific secretome from host-derived proteins in a complex tissue microenvironment in vivo. METHODS In this study, we present the generation and characterization of a novel hiPSC line, L274G-hiPSC, expressing the murine mutant methionyl-tRNA synthetase, L274GMmMetRS, which can be used for tracking the cell specific proteome via biorthogonal non-canonical amino acid tagging (BONCAT). We assessed the trilineage differentiation potential of the L274G-hiPSCs in vitro and in vivo. Furthermore, we assessed the cell-specific proteome labelling in the L274G-hiPSC derived cardiomyocytes (L274G-hiPSC-CMs) in vitro following co-culture with wild type human umbilical vein derived endothelial cells and in vivo post transplantation in murine hearts. RESULTS We demonstrated that the L274G-hiPSCs exhibit typical hiPSC characteristics and that we can efficiently track the cell-specific proteome in their differentiated progenies belonging to the three germ lineages, including L274G-hiPSC-CMs. Finally, we demonstrated cell-specific BONCAT in transplanted L274G-hiPSC-CMs. CONCLUSION The novel L274G-hiPSC line can be used to study the cell-specific proteome of hiPSCs in vitro and in vivo, to delineate mechanisms underlying hiPSC-based cell therapies for a variety of regenerative medicine applications.
Collapse
Affiliation(s)
- Divya Sridharan
- Division of Basic and Translational Sciences, Department of Emergency Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Julie A Dougherty
- Division of Basic and Translational Sciences, Department of Emergency Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Uzair Ahmed
- Division of Basic and Translational Sciences, Department of Emergency Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Shridhar K Sanghvi
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
- Department of Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | - Syed Baseeruddin Alvi
- Division of Basic and Translational Sciences, Department of Emergency Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Ki Ho Park
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Helena Islam
- Division of Basic and Translational Sciences, Department of Emergency Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Sue E Knoblaugh
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Elizabeth D Kirby
- Department of Psychology, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Mahmood Khan
- Division of Basic and Translational Sciences, Department of Emergency Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
26
|
Massidda MW, Ashirov D, Demkov A, Sices A, Baker AB. A Computational Model of Mechanical Stretching of Cultured Cells on a Flexible Membrane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597769. [PMID: 38895285 PMCID: PMC11185657 DOI: 10.1101/2024.06.06.597769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Mechanical forces applied to cells are known to regulate a wide variety of biological processes. Recent studies have supported that mechanical forces can cause nuclear deformation, leading to significant alterations in the gene expression and chromatin landscape of the cell. While the stresses and strains applied to cells is it is often known or controlled experimentally on a macroscopic length scale, it is often unclear what the actual forces and displacements are at the microscopic level of the cell. In this work, we created a model of cell deformation during application of mechanical stretch to cultured cells growth on a flexible membrane. This configuration is commonly used is in experimental studies as a means to apply controlled mechanical strains to adherent cultured cells. The parameters used in the study were used for application of strain to a mesenchymal stem cell stretched on a membrane. computational model was created to simulate the stresses and strains within the cell under a variety of stain amplitudes, waveforms and frequencies of mechanical loading with the range of commonly used experimental systems. The results demonstrate the connection between mechanical loading parameters applied through the flexible membrane and the resulting stresses and strains within the cell and nucleus. Using a viscoelastic model of chromatin, we connected the results provide to a rough model of resulting deformation within chromatin from the forces applied to the nucleus. Overall, the model is useful in providing insight between experimentally applied mechanical forces and the actual forces within the cell to better interpret the results of experimental studies. Statement of Significance In this work, we created a computational model of the mechanical stretching of cell on a flexible membrane under cyclic mechanical loading. This model provides insight into the forces and displacements inside of cell that result from that application of stretch. As many experiments use this set up, our work is relevant to interpreting many studies that use mechanical stretch to stimulate mechanotransduction.
Collapse
|
27
|
Li YX, Wei SQ, Li S, Zheng PS. Strategies and Challenges of Mesenchymal Stem Cells-Derived Extracellular Vesicles in Infertility. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:385-403. [PMID: 38009217 DOI: 10.1089/ten.teb.2023.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
Having genetically related offspring remains an unattainable dream for couples with reproductive failure. Mesenchymal stem cells (MSCs) are multipotent stromal cells derived from various human tissues and organs. As critical paracrine effectors of MSCs, extracellular vesicles (EVs) can carry and deliver bioactive content, thereby participating in intercellular communication and determining cell fate. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have shown promising therapeutic effects, including repairing injured endometria, restoration of ovarian functions, and improving sperm quantity, morphology, and motility, owing to their regenerative potential, abundant sources, high proliferation rates, low immunogenicity, and lack of ethical issues. However, limited knowledge on purification and isolation of MSC-EVs, therapeutic effects, and unpredictable safety have caused challenges in overcoming female and male infertility. To overcome them, future studies should focus on modification/engineering of MSC-EVs with therapeutic biomolecules and combining attractive biomaterials and MSC-EVs. This review highlights the latest studies on MSC-EVs therapies in infertility and the major challenges that must be overcome before clinical translation.
Collapse
Affiliation(s)
- Yuan-Xing Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Si-Qi Wei
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Shan Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Peng-Sheng Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
28
|
Tati V, Mitra S, Basu S, Shukla S. Bone marrow mesenchymal stem cell-derived extracellular vesicles promote corneal epithelial repair and suppress apoptosis via modulation of Caspase-3 in vitro. FEBS Open Bio 2024; 14:968-982. [PMID: 38684330 PMCID: PMC11494918 DOI: 10.1002/2211-5463.13804] [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: 11/03/2023] [Revised: 02/21/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Corneal injuries are the major cause of blindness and visual impairment. Available treatments are limited by their efficacy and side effects. Mesenchymal stem cell-derived extracellular vesicles are presumed as functional equivalents and potential candidates for cell-free therapy. This study reports isolation and characterization of extracellular vesicles from human bone marrow mesenchymal stem cells and evaluates their role in mediating epithelial repair and apoptosis in cultured corneal epithelial cells through scratch assay, PCR, immunofluorescence, and flow cytometry in vitro. The isolated extracellular vesicles were spherical, < 150 nm in diameter, and characterized as CD9+, CD63+, CD81+, TSG101+, and Calnexin-. Further, these vesicles promoted corneal epithelial repair by enhancing proliferation and suppressed apoptosis by regulating the expression of BAD, P53, BCL-2, and cleaved CASPASE-3. Thus, our results suggest that BM-MSC-EVs might have the potential to be used for the treatment of injury-induced corneal epithelial defects. Clinical translation of this work would require further investigations.
Collapse
Affiliation(s)
- Vasudeva Tati
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
| | - Sreya Mitra
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
| | - Sayan Basu
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
- Shantilal Shanghvi Cornea Institute, L V Prasad Eye InstituteHyderabadIndia
| | - Sachin Shukla
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research FoundationL V Prasad Eye InstituteHyderabadIndia
- Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular RegenerationL V Prasad Eye InstituteHyderabadIndia
| |
Collapse
|
29
|
Shao Z, Wang B, Gao H, Zhang S. Microenvironmental interference with intra-articular stem cell regeneration influences the onset and progression of arthritis. Front Genet 2024; 15:1380696. [PMID: 38841721 PMCID: PMC11150611 DOI: 10.3389/fgene.2024.1380696] [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: 02/02/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Studies have indicated that the preservation of joint health and the facilitation of damage recovery are predominantly contingent upon the joint's microenvironment, including cell-cell interactions, the extracellular matrix's composition, and the existence of local growth factors. Mesenchymal stem cells (MSCs), which possess the capacity to self-renew and specialize in many directions, respond to cues from the microenvironment, and aid in the regeneration of bone and cartilage, are crucial to this process. Changes in the microenvironment (such as an increase in inflammatory mediators or the breakdown of the extracellular matrix) in the pathological context of arthritis might interfere with stem cell activation and reduce their ability to regenerate. This paper investigates the potential role of joint microenvironmental variables in promoting or inhibiting the development of arthritis by influencing stem cells' ability to regenerate. The present status of research on stem cell activity in the joint microenvironment is also outlined, and potential directions for developing new treatments for arthritis that make use of these intervention techniques to boost stem cell regenerative potential through altering the intra-articular environment are also investigated. This review's objectives are to investigate these processes, offer fresh perspectives, and offer a solid scientific foundation for the creation of arthritic treatment plans in the future.
Collapse
Affiliation(s)
| | | | | | - Shenqi Zhang
- Department of Joint and Sports Medicine, Zaozhuang Municipal Hospital Affiliated to Jining Medical University, Zaozhuang, Shandong, China
| |
Collapse
|
30
|
Lange M, Babczyk P, Tobiasch E. Exosomes: A New Hope for Angiogenesis-Mediated Bone Regeneration. Int J Mol Sci 2024; 25:5204. [PMID: 38791243 PMCID: PMC11120942 DOI: 10.3390/ijms25105204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Bone is a metabolically dynamic structure that is generally remodeled throughout the lifetime of an individual but often causes problems with increasing age. A key player for bone development and homeostasis, but also under pathological conditions, is the bone vasculature. This complex system of arteries, veins, and capillaries forms distinct structures where each subset of endothelial cells has important functions. Starting with the basic process of angiogenesis and bone-specific blood vessel formation, coupled with initial bone formation, the importance of different vascular structures is highlighted with respect to how these structures are maintained or changed during homeostasis, aging, and pathological conditions. After exemplifying the current knowledge on bone vasculature, this review will move on to exosomes, a novel hotspot of scientific research. Exosomes will be introduced starting from their discovery via current isolation procedures and state-of-the-art characterization to their role in bone vascular development, homeostasis, and bone regeneration and repair while summarizing the underlying signal transduction pathways. With respect to their role in these processes, especially mesenchymal stem cell-derived extracellular vesicles are of interest, which leads to a discussion on patented applications and an update on ongoing clinical trials. Taken together, this review provides an overview of bone vasculature and bone regeneration, with a major focus on how exosomes influence this intricate system, as they might be useful for therapeutic purposes in the near future.
Collapse
Affiliation(s)
- Martin Lange
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Patrick Babczyk
- Department of Natural Sciences, University Bonn-Rhein-Sieg, D-53559 Rheinbach, Germany
| | - Edda Tobiasch
- Department of Natural Sciences, University Bonn-Rhein-Sieg, D-53559 Rheinbach, Germany
| |
Collapse
|
31
|
Adamičková A, Kyselovic J, Adamička M, Chomaničová N, Valášková S, Šalingová B, Molitorisová M, Červenák Z, Danišovič Ľ, Gažová A. Effects of Ibuprofen and Diclofenac Pre-Treatment on Viability and Apoptosis Processes in Human Dental Pulp Stem Cells. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:787. [PMID: 38792973 PMCID: PMC11123081 DOI: 10.3390/medicina60050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: Stem cell-based regeneration strategies have shown therapeutic efficacy in various fields of regenerative medicine. These include bone healing after bone augmentation, often complicated by pain, which is managed by using nonsteroidal anti-inflammatory drugs (NSAIDs). However, information is limited about how NSAIDs affect the therapeutic potential of stem cells. Materials and Methods: We investigated the effects of ibuprofen and diclofenac on the characteristics, morphology, and immunophenotype of human mesenchymal stromal cells isolated from the dental pulp (DPSCs) and cultured in vitro, as well as their effects on the expression of angiogenic growth factors (VEGFA and HGF) and selected genes in apoptosis signalling pathways (BAX, BAK, CASP3, CASP9, and BCL2). Results: Ibuprofen and diclofenac significantly reduced the viability of DPSCs, while the expression of mesenchymal stem cell surface markers was unaffected. Both ibuprofen and diclofenac treatment significantly upregulated the expression of HGF, while the expression of VEGFA remained unchanged. Ibuprofen significantly altered the expression of several apoptosis-related genes, including the upregulation of CASP9 and BCL2, with decreased CASP3 expression. BAK, CASP3, CASP9, and BCL2 expressions were significantly increased in the diclofenac-treated DPSCs, while no difference was demonstrated in BAX expression. Conclusions: Our results suggest that concomitant use of the NSAIDs ibuprofen or diclofenac with stem cell therapy may negatively impact cell viability and alter the expression of apoptosis-related genes, affecting the efficacy of stem cell therapy.
Collapse
Affiliation(s)
- Adriana Adamičková
- 5th Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia
| | - Jan Kyselovic
- 5th Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy, 04181 Košice, Slovakia
| | - Matúš Adamička
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia; (M.A.)
| | - Nikola Chomaničová
- International Laser Centre, Slovak Centre of Scientific and Technical Information, Lamačská cesta 7315/8A, 84104 Bratislava, Slovakia
| | - Simona Valášková
- International Laser Centre, Slovak Centre of Scientific and Technical Information, Lamačská cesta 7315/8A, 84104 Bratislava, Slovakia
| | - Barbara Šalingová
- 5th Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia
| | - Miroslava Molitorisová
- International Laser Centre, Slovak Centre of Scientific and Technical Information, Lamačská cesta 7315/8A, 84104 Bratislava, Slovakia
| | - Zdenko Červenák
- 5th Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia
| | - Ľuboš Danišovič
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia; (M.A.)
| | - Andrea Gažová
- Institute of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Comenius University Bratislava, Špitálska 24, 81372 Bratislava, Slovakia
| |
Collapse
|
32
|
Kurawaki S, Nakashima A, Ishiuchi N, Kanai R, Maeda S, Sasaki K, Masaki T. Mesenchymal stem cells pretreated with interferon-gamma attenuate renal fibrosis by enhancing regulatory T cell induction. Sci Rep 2024; 14:10251. [PMID: 38704512 PMCID: PMC11069572 DOI: 10.1038/s41598-024-60928-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Mesenchymal stem cells (MSCs) exert their anti-inflammatory and anti-fibrotic effects by secreting various humoral factors. Interferon-gamma (IFN-γ) can enhance these effects of MSCs, and enhancement of regulatory T (Treg) cell induction is thought to be an underlying mechanism. However, the extent to which Treg cell induction by MSCs pretreated with IFN-γ (IFN-γ MSCs) ameliorates renal fibrosis remains unknown. In this study, we investigated the effects of Treg cell induction by IFN-γ MSCs on renal inflammation and fibrosis using an siRNA knockdown system. Administration of IFN-γ MSCs induced Treg cells and inhibited infiltration of inflammatory cells in ischemia reperfusion injury (IRI) rats more drastically than control MSCs without IFN-γ pretreatment. In addition, administration of IFN-γ MSCs more significantly attenuated renal fibrosis compared with control MSCs. Indoleamine 2,3-dioxygenase (IDO) expression levels in conditioned medium from MSCs were enhanced by IFN-γ pretreatment. Moreover, IDO1 knockdown in IFN-γ MSCs reduced their anti-inflammatory and anti-fibrotic effects in IRI rats by reducing Treg cell induction. Our findings suggest that the increase of Treg cells induced by enhanced secretion of IDO by IFN-γ MSCs played a pivotal role in their anti-fibrotic effects. Administration of IFN-γ MSCs may potentially be a useful therapy to prevent renal fibrosis progression.
Collapse
Affiliation(s)
- So Kurawaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ryo Kanai
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Satoshi Maeda
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
33
|
Ramzan F, Khalid S, Ekram S, Salim A, Frazier T, Begum S, Mohiuddin OA, Khan I. 3D bio scaffold support osteogenic differentiation of mesenchymal stem cells. Cell Biol Int 2024; 48:594-609. [PMID: 38321826 DOI: 10.1002/cbin.12131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024]
Abstract
The regeneration of osteochondral lesions by tissue engineering techniques is challenging due to the lack of physicochemical characteristics and dual-lineage (osteogenesis and chondrogenesis). A scaffold with better mechanical properties and dual lineage capability is required for the regeneration of osteochondral defects. In this study, a hydrogel prepared from decellularized human umbilical cord tissue was developed and evaluated for osteochondral regeneration. Mesenchymal stem cells (MSCs) isolated from the umbilical cord were seeded with hydrogel for 28 days, and cell-hydrogel composites were cultured in basal and osteogenic media. Alizarin red staining, quantitative polymerase chain reaction, and immunofluorescent staining were used to confirm that the hydrogel was biocompatible and capable of inducing osteogenic differentiation in umbilical cord-derived MSCs. The findings demonstrate that human MSCs differentiated into an osteogenic lineage following 28 days of cultivation in basal and osteoinductive media. The expression was higher in the cell-hydrogel composites cultured in osteoinductive media, as evidenced by increased levels of messenger RNA and protein expression of osteogenic markers as compared to basal media cultured cell-hydrogel composites. Additionally, calcium deposits were also observed, which provide additional evidence of osteogenic differentiation. The findings demonstrate that the hydrogel is biocompatible with MSCs and possesses osteoinductive capability in vitro. It may be potentially useful for osteochondral regeneration.
Collapse
Affiliation(s)
- Faiza Ramzan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shumaila Khalid
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Sobia Ekram
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Sumreen Begum
- Stem Cell Research Laboratory (SCRL), Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Omair A Mohiuddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
34
|
Jeong SY, Park BW, Kim J, Lee S, You H, Lee J, Lee S, Park JH, Kim J, Sim W, Ban K, Park J, Park HJ, Kim S. Hyaluronic acid stimulation of stem cells for cardiac repair: a cell-free strategy for myocardial infarct. J Nanobiotechnology 2024; 22:149. [PMID: 38570846 PMCID: PMC10993512 DOI: 10.1186/s12951-024-02410-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI), a representative form of ischemic heart disease, remains a huge burden worldwide. This study aimed to explore whether extracellular vesicles (EVs) secreted from hyaluronic acid (HA)-primed induced mesenchymal stem cells (HA-iMSC-EVs) could enhance the cardiac repair after MI. RESULTS HA-iMSC-EVs showed typical characteristics for EVs such as morphology, size, and marker proteins expression. Compared with iMSC-EVs, HA-iMSC-EVs showed enhanced tube formation and survival against oxidative stress in endothelial cells, while reduced reactive oxygen species (ROS) generation in cardiomyocytes. In THP-1 macrophages, both types of EVs markedly reduced the expression of pro-inflammatory signaling players, whereas HA-iMSC-EVs were more potent in augmenting anti-inflammatory markers. A significant decrease of inflammasome proteins was observed in HA-iMSC-EV-treated THP-1. Further, phospho-SMAD2 as well as fibrosis markers in TGF-β1-stimulated cardiomyocytes were reduced in HA-iMSC-EVs treatment. Proteomic data showed that HA-iMSC-EVs were enriched with multiple pathways including immunity, extracellular matrix organization, angiogenesis, and cell cycle. The localization of HA-iMSC-EVs in myocardium was confirmed after delivery by either intravenous or intramyocardial route, with the latter increased intensity. Echocardiography revealed that intramyocardial HA-iMSC-EVs injections improved cardiac function and reduced adverse cardiac remodeling and necrotic size in MI heart. Histologically, MI hearts receiving HA-iMSC-EVs had increased capillary density and viable myocardium, while showed reduced fibrosis. CONCLUSIONS Our results suggest that HA-iMSC-EVs improve cardiac function by augmenting vessel growth, while reducing ROS generation, inflammation, and fibrosis in MI heart.
Collapse
Affiliation(s)
- Seon-Yeong Jeong
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
- Catholic High-Performance Cell Therapy Center and Department of Medical Life Science, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Seulki Lee
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Haedeun You
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Joohyun Lee
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Susie Lee
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Jinju Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Woosup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Joonghoon Park
- Graduate School of International Agricultural Technology, Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, 25354, South Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea.
- Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea.
| |
Collapse
|
35
|
Chen F, Che Z, Liu Y, Luo P, Xiao L, Song Y, Wang C, Dong Z, Li M, Tipoe GL, Yang M, Lv Y, Zhang H, Wang F, Xiao J. Invigorating human MSCs for transplantation therapy via Nrf2/DKK1 co-stimulation in an acute-on-chronic liver failure mouse model. Gastroenterol Rep (Oxf) 2024; 12:goae016. [PMID: 38529014 PMCID: PMC10963075 DOI: 10.1093/gastro/goae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/27/2023] [Accepted: 02/21/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Since boosting stem cell resilience in stressful environments is critical for the therapeutic efficacy of stem cell-based transplantations in liver disease, this study aimed to establish the efficacy of a transient plasmid-based preconditioning strategy for boosting the capability of mesenchymal stromal cells (MSCs) for anti-inflammation/antioxidant defenses and paracrine actions in recipient hepatocytes. METHODS Human adipose mesenchymal stem cells (hADMSCs) were subjected to transfer, either with or without the nuclear factor erythroid 2-related factor 2 (Nrf2)/Dickkopf1 (DKK1) genes, followed by exposure to TNF-α/H2O2. Mouse models were subjected to acute chronic liver failure (ACLF) and subsequently injected with either transfected or untransfected MSCs. These hADMSCs and ACLF mouse models were used to investigate the interaction between Nrf2/DKK1 and the hepatocyte receptor cytoskeleton-associated protein 4 (CKAP4). RESULTS Activation of Nrf2 and DKK1 enhanced the anti-stress capacity of MSCs in vitro. In a murine model of ACLF, transient co-overexpression of Nrf2 and DKK1 via plasmid transfection improved MSC resilience against inflammatory and oxidative assaults, boosted MSC transplantation efficacy, and promoted recipient liver regeneration due to a shift from the activation of the anti-regenerative IFN-γ/STAT1 pathway to the pro-regenerative IL-6/STAT3 pathway in the liver. Importantly, the therapeutic benefits of MSC transplantation were nullified when the receptor CKAP4, which interacts with DKK1, was specifically removed from recipient hepatocytes. However, the removal of the another receptor low-density lipoprotein receptor-related protein 6 (LRP6) had no impact on the effectiveness of MSC transplantation. Moreover, in long-term observations, no tumorigenicity was detected in mice following transplantation of transiently preconditioned MSCs. CONCLUSIONS Co-stimulation with Nrf2/DKK1 safely improved the efficacy of human MSC-based therapies in murine models of ACLF through CKAP4-dependent paracrine mechanisms.
Collapse
Affiliation(s)
- Feng Chen
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Yingxia Liu
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - Pingping Luo
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Lu Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Yali Song
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Cunchuan Wang
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Zhiyong Dong
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Mianhuan Li
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - George L Tipoe
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Min Yang
- National Clinical Research Center for Infectious Diseases, Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, P. R. China
| | - Yi Lv
- Laboratory of Neuroendocrinology, Fujian Key Laboratory of Developmental and Neurobiology, School of Life Sciences, Fujian Normal University, Fuzhou, Fujian, P. R. China
| | - Hong Zhang
- Department of Surgery, The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, Guangdong, P. R. China
| | - Fei Wang
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, P. R. China
- Department of Surgery, The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, Guangdong, P. R. China
| |
Collapse
|
36
|
Zakaria MF, Sonoda S, Kato H, Ma L, Uehara N, Kyumoto-Nakamura Y, Sharifa MM, Yu L, Dai L, Yamauchi-Tomoda E, Aijima R, Yamaza H, Nishimura F, Yamaza T. Erythropoietin receptor signal is crucial for periodontal ligament stem cell-based tissue reconstruction in periodontal disease. Sci Rep 2024; 14:6719. [PMID: 38509204 PMCID: PMC10954634 DOI: 10.1038/s41598-024-57361-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/18/2024] [Indexed: 03/22/2024] Open
Abstract
Alveolar bone loss caused by periodontal disease eventually leads to tooth loss. Periodontal ligament stem cells (PDLSCs) are the tissue-specific cells for maintaining and repairing the periodontal ligament, cementum, and alveolar bone. Here, we investigated the role of erythropoietin receptor (EPOR), which regulates the microenvironment-modulating function of mesenchymal stem cells, in PDLSC-based periodontal therapy. We isolated PDLSCs from patients with chronic periodontal disease and healthy donors, referred to as PD-PDLSCs and Cont-PDLSCs, respectively. PD-PDLSCs exhibited reduced potency of periodontal tissue regeneration and lower expression of EPOR compared to Cont-PDLSCs. EPOR-silencing suppressed the potency of Cont-PDLSCs mimicking PD-PDLSCs, whereas EPO-mediated EPOR activation rejuvenated the reduced potency of PD-PDLSCs. Furthermore, we locally transplanted EPOR-silenced and EPOR-activated PDLSCs into the gingiva around the teeth of ligament-induced periodontitis model mice and demonstrated that EPOR in PDLSCs participated in the regeneration of the periodontal ligament, cementum, and alveolar bone in the ligated teeth. The EPOR-mediated paracrine function of PDLSCs maintains periodontal immune suppression and bone metabolic balance via osteoclasts and osteoblasts in the periodontitis model mice. Taken together, these results suggest that EPOR signaling is crucial for PDLSC-based periodontal regeneration and paves the way for the development of novel options for periodontal therapy.
Collapse
Affiliation(s)
- Mhd Fouad Zakaria
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of Periodontology, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Lan Ma
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Guangdong Provincial Key Laboratory of Stomatology, South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yukari Kyumoto-Nakamura
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - M Majd Sharifa
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Liting Yu
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Lisha Dai
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Erika Yamauchi-Tomoda
- Department of Oral and Maxillofacial Radiology, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Reona Aijima
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Fusanori Nishimura
- Department of Periodontology, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
37
|
Seok J, Park HS, Cetin E, Ghasroldasht MM, Liakath FB, Al-Hendy A. The potent paracrine effect of umbilical cord mesenchymal stem cells mediates mitochondrial quality control to restore chemotherapy-induced damage in ovarian granulosa cells. Biomed Pharmacother 2024; 172:116263. [PMID: 38350369 DOI: 10.1016/j.biopha.2024.116263] [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: 12/08/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
The basic principle of chemotherapy is to attack cells with fast growth, and cancer cells are targeted by anticancer drugs because they have a faster growth rate than normal cells. High doses of anticancer drugs may cause an irreversible decline in reproductive capacity, and novel approaches for fertility preservation and/or restoration after anticancer treatment are urgently needed. Here, we provide important insights into the recovery of human reproductive cells damaged by chemotherapy. We performed a detailed screening of the cytokines of various human mesenchymal stem cells (hMSCs) to select superior MSCs. Also, we analyzed the Ovarian granulosa cell (OGC)-)-specific functions for restoring function, apoptosis, and mitochondrial functions to confirm the recovery mechanism in damaged OGCs. As a result, we demonstrated that conditioned media (CM) of Umbilical cord mesenchymal stem cells (UC-MSCs) could restore the functions of damaged OGCs primarily through antiapoptotic and antioxidant effects. Furthermore, CM changed the phenotype of damaged OGCs to an energetic status by restoring mitochondrial function and enhanced the mitochondrial metabolic activity decreased by chemotherapy. Finally, we demonstrated that the restoration of mitochondrial function in damaged OGCs was mediated through mitochondrial autophagy (mitophagy). Our findings offer new insights into the potential of stem cell-based therapy for fertility preservation and/or restoration in female cancer patients.
Collapse
Affiliation(s)
- Jin Seok
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Hang-Soo Park
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Esra Cetin
- Department of Obstetrics and Gynecology, Hurley Medical Center, Michigan State University, 1 Hurley Plaza, Flint, MI 48503, USA
| | | | - Farzana Begum Liakath
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, 5841 A. Maryland Ave, Chicago, IL 60637, USA.
| |
Collapse
|
38
|
Huang G, Shen H, Xu K, Shen Y, Jiale Jin, Chu G, Xing H, Feng Z, Wang Y. Single-Cell Microgel Encapsulation Improves the Therapeutic Efficacy of Mesenchymal Stem Cells in Treating Intervertebral Disc Degeneration via Inhibiting Pyroptosis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0311. [PMID: 38371273 PMCID: PMC10871001 DOI: 10.34133/research.0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/14/2024] [Indexed: 02/20/2024]
Abstract
While mesenchymal stem cell (MSC) shows great potentials in treating intervertebral disc degeneration, most MSC die soon after intradiscal transplantation, resulting in inferior therapeutic efficacy. Currently, bulk hydrogels are the common solution to improve MSC survival in tissues, although hydrogel encapsulation impairs MSC migration and disrupts extracellular microenvironment. Cell hydrogel encapsulation has been proposed to overcome the limitation of traditional bulk hydrogels, yet this technique has not been used in treating disc degeneration. Using a layer-by-layer self-assembly technique, we fabricated alginate and gelatin microgel to encapsulate individual MSC for treating disc degeneration. The small size of microgel allowed intradiscal injection of coated MSC. We demonstrated that pyroptosis was involved in MSC death under oxidative stress stimulation, and microgel coating suppressed pyroptosis activation by maintaining mitochondria homeostasis. Microgel coating protected MSC in the harsh disc microenvironment, while retaining vital cellular functions such as migration, proliferation, and differentiation. In a rat model of disc degeneration, coated MSC exhibits prolonged retention in the disc and better efficacy of attenuating disc degeneration, as compared with bare MSC treatment alone. Further, microgel-coated MSC exhibited improved therapeutic effects in treating disc degeneration via suppressing the activation of pyroptosis in the disc. For the first time, microgel-encapsulated MSC was used to treat disc degeneration and obtain encouraging outcomes. The developed biocompatible single-cell hydrogel is an effective strategy to protect MSC and maintain cellular functions and may be an efficacious approach to improving the efficacy of MSC therapy in treating disc degeneration. The objective of this study is to improve the efficacy of cell therapy for treating disc degeneration using single-cell hydrogel encapsulation and further to understand related cytoprotective mechanisms.
Collapse
Affiliation(s)
- Guanrui Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haotian Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kaiwang Xu
- Zhejiang University, Hangzhou 310058, China
| | - Yifan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiale Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Guangyu Chu
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hongyuan Xing
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhiyun Feng
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yue Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
39
|
Gao M, Guo H, Dong X, Wang Z, Yang Z, Shang Q, Wang Q. Regulation of inflammation during wound healing: the function of mesenchymal stem cells and strategies for therapeutic enhancement. Front Pharmacol 2024; 15:1345779. [PMID: 38425646 PMCID: PMC10901993 DOI: 10.3389/fphar.2024.1345779] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
A wound takes a long time to heal and involves several steps. Following tissue injury, inflammation is the primary cause of tissue regeneration and repair processes. As a result, the pathophysiological processes involving skin damage, healing, and remodeling depend critically on the control of inflammation. The fact that it is a feasible target for improving the prognosis of wound healing has lately become clear. Mesenchymal stem cells (MSCs) are an innovative and effective therapeutic option for wound healing due to their immunomodulatory and paracrine properties. By controlling the inflammatory milieu of wounds through immunomodulation, transplanted MSCs have been shown to speed up the healing process. In addition to other immunomodulatory mechanisms, including handling neutrophil activity and modifying macrophage polarization, there may be modifications to the activation of T cells, natural killer (NK) cells, and dendritic cells (DCs). Furthermore, several studies have shown that pretreating MSCs improves their ability to modulate immunity. In this review, we summarize the existing knowledge about how MSCs influence local inflammation in wounds by influencing immunity to facilitate the healing process. We also provide an overview of MSCs optimizing techniques when used to treat wounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qiying Wang
- Department of Plastic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
40
|
Walton BL, Shattuck-Brandt R, Hamann CA, Tung VW, Colazo JM, Brand DD, Hasty KA, Duvall CL, Brunger JM. A programmable arthritis-specific receptor for guided articular cartilage regenerative medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578281. [PMID: 38352576 PMCID: PMC10862827 DOI: 10.1101/2024.01.31.578281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Objective Investigational cell therapies have been developed as disease-modifying agents for the treatment of osteoarthritis (OA), including those that inducibly respond to inflammatory factors driving OA progression. However, dysregulated inflammatory cascades do not specifically signify the presence of OA. Here, we deploy a synthetic receptor platform that regulates cell behaviors in an arthritis-specific fashion to confine transgene expression to sites characterized by cartilage degeneration. Methods An scFv specific for type II collagen (CII) was used to produce a synthetic Notch (synNotch) receptor that enables "CII-synNotch" mesenchymal stromal cells (MSCs) to recognize CII fibers exposed in damaged cartilage. Engineered cell activation by both CII-treated culture surfaces and on primary tissue samples was measured via inducible reporter transgene expression. TGFβ3-expressing cells were assessed for cartilage anabolic gene expression via qRT-PCR. In a co-culture with CII-synNotch MSCs engineered to express IL-1Ra, ATDC5 chondrocytes were stimulated with IL-1α, and inflammatory responses of ATDC5s were profiled via qRT-PCR and an NF-κB reporter assay. Results CII-synNotch MSCs are highly responsive to CII, displaying activation ranges over 40-fold in response to physiologic CII inputs. CII-synNotch cells exhibit the capacity to distinguish between healthy and damaged cartilage tissue and constrain transgene expression to regions of exposed CII fibers. Receptor-regulated TGFβ3 expression resulted in upregulation of Acan and Col2a1 in MSCs, and inducible IL-1Ra expression by engineered CII-synNotch MSCs reduced pro-inflammatory gene expression in chondrocytes. Conclusion This work demonstrates proof-of-concept that the synNotch platform guides MSCs for spatially regulated, disease-dependent delivery of OA-relevant biologic drugs.
Collapse
Affiliation(s)
- Bonnie L. Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | | | - Catherine A. Hamann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Victoria W. Tung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - David D. Brand
- Research Service, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Karen A. Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis VA Medical Center, Memphis, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA
| |
Collapse
|
41
|
Somadder R, Faraj L, Datta S, Kanapathipillai M, Ghosh G. Effect of extracellular matrices on production and potency of mesenchymal stem cell-derived exosomes. Biotechnol J 2024; 19:e2300474. [PMID: 38403471 DOI: 10.1002/biot.202300474] [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: 09/10/2023] [Revised: 12/17/2023] [Accepted: 12/23/2023] [Indexed: 02/27/2024]
Abstract
Mesenchymal stem cell (MSC) derived exosomes have emerged as potential acellular therapeutics for various tissue regenerative applications. However, successful clinical translation of exosome-based therapy is limited by lack of a structured production platform. Thus, in this study, the effect of decellularized extracellular matrix (dECM) was assessed on the production and potency of exosomes secreted by bone marrow-derived human MSCs. The results indicate that there was a ∼2-fold increase in MSC-exosome production when MSCs were cultured on dECM compared to TCP. Further, our study revealed that dECM generation induced by ascorbic acid (AA) up to 100 µg mL-1 highly increased exosome yield thereby indicating a potential scale up method for MSC exosome production. The bioactivity of exosomes was investigated by their ability to improve the healing of wounded human skin explants. Wound closure was enhanced in the presence of exosomes isolated from MSCs cultured on ascorbic acid-induced dECM compared to TCP generated MSC-exosomes. In summary, this study suggests a promising solution to a major bottleneck in large-scale production of MSC exosomes for cell-free therapy.
Collapse
Affiliation(s)
- Rittika Somadder
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Lina Faraj
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Saurav Datta
- Amgen Bioprocessing Center, Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, USA
| | - Mathumai Kanapathipillai
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Gargi Ghosh
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
- Amgen Bioprocessing Center, Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, USA
| |
Collapse
|
42
|
Xiong J, Wang F, Yang Y, Yang Y, Liu Z. Preventive effect of human umbilical cord mesenchymal stem cells on skin aging in rats. Heliyon 2024; 10:e24342. [PMID: 38293418 PMCID: PMC10826728 DOI: 10.1016/j.heliyon.2024.e24342] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/21/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
The irreversibility of aging makes anti-aging become an important research direction in the field of medical research. As the most direct manifestation of human aging, skin aging has been paid more and more attention. Stem cells have been used as a basis for anti-aging studies in skin, of which adipose-derived mesenchymal stem cells are more commonly used. In this study, human umbilical cord mesenchymal stem cells were used, and human umbilical cord mesenchymal stem cells were intervened while making a skin aging model, which was planned to reduce the process of preventing skin aging in the study method. At the end of the experiment, rat skin and serum were taken for relevant data detection. The results showed that the contents of EGF and VEGF in serum and skin tissue of rats increased and the content of MDA decreased after the application of human umbilical cord mesenchymal stem cells. At the same time, hUCMSC intervention increased skin thickness, increased dermal vessels, increased type I collagen type III collagen mRNA expression, and decreased MMP-1 content in rats. The results showed that hUCMSC could prevent skin aging in rats.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Plastic and Burns, The Affiliated Hospital of Guizhou Medical University, China
- Department of Plastic and Burns, First Affiliated Hospital of the National Sun Yat-sen University, Guizhou Hospital, China
| | - Fan Wang
- Department of Plastic and Burns, The Affiliated Hospital of Guizhou Medical University, China
| | - Yutong Yang
- Department of Plastic and Burns, The Affiliated Hospital of Guizhou Medical University, China
| | - Yuxi Yang
- Department of Plastic and Burns, The Affiliated Hospital of Guizhou Medical University, China
| | - Zhongshan Liu
- Department of Plastic and Burns, The Affiliated Hospital of Guizhou Medical University, China
| |
Collapse
|
43
|
Li XH, Huang P, Cheng HP, Zhou Y, Feng DD, Yue SJ, Han Y, Luo ZQ. NMDAR activation attenuates the protective effect of BM-MSCs on bleomycin-induced ALI via the COX-2/PGE 2 pathway. Heliyon 2024; 10:e23723. [PMID: 38205313 PMCID: PMC10776937 DOI: 10.1016/j.heliyon.2023.e23723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
N-methyl-d-aspartate (NMDA) receptor (NMDAR) activation mediates glutamate (Glu) toxicity and involves bleomycin (BLM)-induced acute lung injury (ALI). We have reported that bone marrow-derived mesenchymal stem cells (BM-MSCs) are NMDAR-regulated target cells, and NMDAR activation inhibits the protective effect of BM-MSCs on BLM-induced pulmonary fibrosis, but its effect on ALI remains unknown. Here, we found that Glu release was significantly elevated in plasma of mice at d 7 after intratracheally injected with BLM. BM-MSCs were pretreated with NMDA (the selective agonist of NMDAR) and transplanted into the recipient mice after the BLM challenge. BM-MSCs administration significantly alleviated the pathological changes, inflammatory response, myeloperoxidase activity, and malondialdehyde content in the damaged lungs, but NMDA-pretreated BM-MSCs did not ameliorate BLM-induced lung injury in vivo. Moreover, NMDA down-regulated prostaglandin E2 (PGE2) secretion and cyclooxygenase (COX)-2 expression instead of COX-1 expression in BM-MSCs in vitro. We also found that NMDAR1 expression was increased and COX-2 expression was decreased, but COX-1 expression was not changed in primary BM-MSCs of BLM-induced ALI mice. Further, the cultured supernatants of lipopolysaccharide (LPS)-pretreated RAW264.7 macrophages were collected to detect inflammatory factors after co-culture with NMDA-pretreated BM-MSCs. The co-culture experiments showed that NMDA precondition inhibited the anti-inflammatory effect of BM-MSCs on LPS-induced macrophage inflammation, and PGE2 could partially alleviate this inhibition. Our findings suggest that NMDAR activation attenuated the protective effect of BM-MSCs on BLM-induced ALI in vivo. NMDAR activation inhibited COX-2 expression and PGE2 secretion in BM-MSCs and weakened the anti-inflammatory effect of BM-MSCs on LPS-induced macrophage inflammation in vitro. In conclusion, NMDAR activation attenuates the protective effect of BM-MSCs on BLM-induced ALI via the COX-2/PGE2 pathway. Keywords: Acute Lung Injury, BM-MSCs, NMDA receptor, COX-1/2, PGE2.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Pu Huang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
- Health Management Center, Changsha Central Hospital Affiliated to Nanhua University, Changsha, 410018, China
| | - Hai-Peng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Dan-Dan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Shao-Jie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yang Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, 410078, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, 410078, China
| |
Collapse
|
44
|
Guo C, Cui Y, Jiao M, Yao J, Zhao J, Tian Y, Dong J, Liao L. Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Front Endocrinol (Lausanne) 2024; 14:1256375. [PMID: 38260142 PMCID: PMC10801024 DOI: 10.3389/fendo.2023.1256375] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
The energy needs of tubular epithelial components, especially proximal tubular epithelial cells (PTECs), are high and they heavily depend on aerobic metabolism. As a result, they are particularly vulnerable to various injuries caused by factors such as ischemia, proteinuria, toxins, and elevated glucose levels. Initial metabolic and phenotypic changes in PTECs after injury are likely an attempt at survival and repair. Nevertheless, in cases of recurrent or prolonged injury, PTECs have the potential to undergo a transition to a secretory state, leading to the generation and discharge of diverse bioactive substances, including transforming growth factor-β, Wnt ligands, hepatocyte growth factor, interleukin (IL)-1β, lactic acid, exosomes, and extracellular vesicles. By promoting fibroblast activation, macrophage recruitment, and endothelial cell loss, these bioactive compounds stimulate communication between epithelial cells and other interstitial cells, ultimately worsening renal damage. This review provides a summary of the latest findings on bioactive compounds that facilitate the communication between these cellular categories, ultimately leading to the advancement of tubulointerstitial fibrosis (TIF).
Collapse
Affiliation(s)
- Congcong Guo
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuying Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jinming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Junyu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yutian Tian
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| |
Collapse
|
45
|
Adamickova A, Chomanicova N, Adamicka M, Valaskova S, Gazova A, Kyselovic J. Isolation and Characterization of Human Dental Pulp Stem Cells Derived from Dental Pulp of Permanent Teeth. Methods Mol Biol 2024; 2835:49-57. [PMID: 39105905 DOI: 10.1007/978-1-0716-3995-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Dental pulp stem cells (DPSCs) are a promising alternative to the source of mesenchymal stem cells (MSCs), as they are readily available in minimally invasive procedures compared to more invasive methods associated with harvesting other MSCs sources. Despite the encouraging pre-clinical outcomes in animal disease models, culture-expanding procedures are needed to obtain a sufficient number of MSCs required for delivery to the damaged site. However, this contributes to increasing regulatory difficulties in translating stem cells and tissue engineering therapy to clinical use. Moreover, discussions continue as to which isolation method is to be preferred when obtaining DPSCs from extracted molars. This protocol describes a simple explant isolation technique of human dental pulp stem cells from the dental pulp of permanent teeth based upon the plastic adherence of MSCs and subsequent outgrowth of cells out of tissue fragments with high efficacy.
Collapse
Affiliation(s)
- Adriana Adamickova
- Faculty of Medicine, 5th Department of Internal Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Nikola Chomanicova
- International Laser Center, Slovak Centre of Scientifc and Technical Information, Bratislava, Slovakia
| | - Matus Adamicka
- Faculty of Medicine, Institute of Medical Biology, Genetics and Clinical Genetics, Comenius University Bratislava, Bratislava, Slovakia
| | - Simona Valaskova
- International Laser Center, Slovak Centre of Scientifc and Technical Information, Bratislava, Slovakia
| | - Andrea Gazova
- Faculty of Medicine, Institute of Pharmacology and Clinical Pharmacology, Comenius University Bratislava, Bratislava, Slovakia
| | - Jan Kyselovic
- Faculty of Medicine, 5th Department of Internal Medicine, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
46
|
Yasan GT, Gunel-Ozcan A. Hypoxia and Hypoxia Mimetic Agents As Potential Priming Approaches to Empower Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:33-54. [PMID: 36642875 DOI: 10.2174/1574888x18666230113143234] [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: 07/29/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSC) exhibit self-renewal capacity and multilineage differentiation potential, making them attractive for research and clinical application. The properties of MSC can vary depending on specific micro-environmental factors. MSC resides in specific niches with low oxygen concentrations, where oxygen functions as a metabolic substrate and a signaling molecule. Conventional physical incubators or chemically hypoxia mimetic agents are applied in cultures to mimic the original low oxygen tension settings where MSC originated. This review aims to focus on the current knowledge of the effects of various physical hypoxic conditions and widely used hypoxia-mimetic agents-PHD inhibitors on mesenchymal stem cells at a cellular and molecular level, including proliferation, stemness, differentiation, viability, apoptosis, senescence, migration, immunomodulation behaviors, as well as epigenetic changes.
Collapse
Affiliation(s)
| | - Aysen Gunel-Ozcan
- Department of Stem Cell Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
47
|
Wang Y, She S, Li W, Zhu J, Li X, Yang F, Dai K. Inhibition of cGAS-STING pathway by stress granules after activation of M2 macrophages by human mesenchymal stem cells against drug induced liver injury. Mol Immunol 2024; 165:42-54. [PMID: 38150981 DOI: 10.1016/j.molimm.2023.12.005] [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: 07/08/2023] [Revised: 11/05/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Cells can produce stress granules (SGs) to protect itself from damage under stress. The cGAS-STING pathway is one of the important pattern recognition pathways in the natural immune system. This study was investigated whether human mesenchymal stem cells (hMSCs) could protect the liver by inducing M2 macrophages to produce SGs during acute drug induced liver injury (DILI) induced by acetaminophen (APAP). METHODS After intragastric administration of APAP in vivo to induce DILI mice model, hMSCs were injected into the tail vein. The co-culture system of hMSCs and M2 macrophages was established in vitro. It was further use SGs inhibitor anisomicin to intervene M2 macrophages. The liver histopathology, liver function, reactive oxygen species (ROS) level, apoptosis pathway, endoplasmic reticulum stress (ERS) level, SGs markers (G3BP1/TIA-1), cGAS-STING pathway, TNF-α, IL-6, IL-1β mRNA levels in liver tissue and M2 macrophages were observed. RESULTS In vivo experiments, it showed that hMSCs could alleviate liver injury, inhibite the level of ROS, apoptosis and ERS, protect liver function in DILI mice. The mount of M2 was increased in the liver. hMSCs could also induce the production of SGs, inhibit the cGAS-STING pathway and reduce TNF-α, IL-6, IL-1β mRNA expression. The results in vitro showed that hMSCs could induce the production of SGs in macrophages, inhibit the cGAS-STING pathway, promote the secretion of IL-4 and IL-13, and reduce TNF-α, IL-6, IL-1β mRNA level in cells. In the process of IL-4 inducing M2 macrophage activation, anisomycin could inhibit the production of SGs, activate the cGAS-STING pathway, and promote the inflammatory factor TNF-α, IL-6, IL-1β mRNA expression in cells. CONCLUSIONS HMSCs had a protective effect on acute DILI in mice induced by APAP. Its mechanism might involve in activating M2 type macrophages, promoting the production of SGs, inhibiting the cGAS-STING pathway, and reducing the expression of pro-inflammatory factors in macrophages, to reduce hepatocytes damage.
Collapse
Affiliation(s)
- Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sha She
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiling Zhu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xun Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Yang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Kai Dai
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
48
|
Cavallero S, Dekali S, Guitard N, Théry H, Hélissey C, François S. Effects of preconditioning with TNFα and IFNγ in angiogenic potential of mesenchymal stromal cell-derived extracellular vesicles. Front Cell Dev Biol 2023; 11:1291016. [PMID: 38178868 PMCID: PMC10764498 DOI: 10.3389/fcell.2023.1291016] [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: 09/08/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction: Mesenchymal stromal cells (MSCs) have demonstrated therapeutic properties both in vitro and in vivo to treat various diseases, including anti-inflammatory, immunomodulatory and pro-angiogenic effects. These therapeutic effects are mediated by their secretome composed of soluble factors and extracellular vesicles (EVs). The composition of EVs reflects the molecular and functional characteristics of parental cells. MSC preconditioning can alter the composition of EVs, thereby influencing their therapeutic potential. Methods: MSCs were subjected to preconditioning with two cytokines, TNFα and IFNγ. Following 24 h of preconditioning, MSC-EVs secreted into the culture supernatant were isolated through tangential filtration. Particle concentration and size distribution were measured by nanoparticle tracking analysis, and the surface antigen expression of the EV-specific CD63 was quantified via Enzyme Linked ImmunoSorbent Assay. The angiogenic potential of MSCEVs obtained after preconditioning MSCs was assessed by the analysis of their protein composition and their influence on human umbilical vein endothelial cell (HUVECs) proliferation, migration, and tube-forming ability. Results: Preconditioning with TNFα and IFNγ did not influence the MSC-EV profile but did induce changes in their protein content. Indeed, the expression of pro-angiogenic proteins increased in EVs from preconditioned MSCs compared to EVs from no-preconditioned MSCs. EVs from preconditioned MSCs tend to stimulate HUVEC migration, proliferation and tubeforming ability. These observations imply the presence of a pro-angiogenic potential in EVs obtained after preconditioning of MSCs with TNFα and IFNγ. Discussion: In conclusion, it appears that the pro-angiogenic potential of EVs is enhanced through preconditioning of MSCs with TNFα and IFNγ. The use of these MSCs-EVs in therapy would circumvent the limitations of current cell-based therapies. Indeed, the therapeutic potential of MSC-EVs presents an attractive strategy for exploiting the clinical benefits of MSC therapy. For example, in the field of regenerative medicine, the exploitation of cell-free therapy using highly pro-angiogenic MSC-EVs is of great interest.
Collapse
Affiliation(s)
- Sophie Cavallero
- Armed Forces Biomedical Research Institute (IRBA), Department of Biological Effects of Radiation, Radiobiology Unit, Brétigny-sur-Orge, France
| | - Samir Dekali
- Armed Forces Biomedical Research Institute (IRBA), Department of Biological Effects of Radiation, Emerging Technologies Risk Unit, Brétigny-sur-Orge, France
| | - Nathalie Guitard
- Armed Forces Biomedical Research Institute (IRBA), Department of Biological Effects of Radiation, Radiobiology Unit, Brétigny-sur-Orge, France
| | - Héléne Théry
- Armed Forces Biomedical Research Institute (IRBA), Department of Biological Effects of Radiation, Radiobiology Unit, Brétigny-sur-Orge, France
| | - Carole Hélissey
- Armed Forces Biomedical Research Institute (IRBA), Department of Biological Effects of Radiation, Radiobiology Unit, Brétigny-sur-Orge, France
- Clinical Unit Research, HIA Begin, Paris, France
| | - Sabine François
- Armed Forces Biomedical Research Institute (IRBA), Department of Biological Effects of Radiation, Radiobiology Unit, Brétigny-sur-Orge, France
| |
Collapse
|
49
|
Alves DVL, Claudio-da-Silva C, Souza MCA, Pinho RT, da Silva WS, Sousa-Vasconcelos PS, Borojevic R, Nogueira CM, Dutra HDS, Takiya CM, Bonfim DC, Rossi MID. Adipose Tissue-Derived Mesenchymal Stromal Cells from Ex-Morbidly Obese Individuals Instruct Macrophages towards a M2-Like Profile In Vitro. Int J Stem Cells 2023; 16:425-437. [PMID: 37643763 PMCID: PMC10686802 DOI: 10.15283/ijsc22172] [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: 10/12/2022] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 08/31/2023] Open
Abstract
Obesity, which continues to increase worldwide, was shown to irreversibly impair the differentiation potential and angiogenic properties of adipose tissue mesenchymal stromal cells (ADSCs). Because these cells are intended for regenerative medicine, especially for the treatment of inflammatory conditions, and the effects of obesity on the immunomodulatory properties of ADSCs are not yet clear, here we investigated how ADSCs isolated from former obese subjects (Ex-Ob) would influence macrophage differentiation and polarization, since these cells are the main instructors of inflammatory responses. Analysis of the subcutaneous adipose tissue (SAT) of overweight (OW) and Ex-Ob subjects showed the maintenance of approximately twice as many macrophages in Ex-Ob SAT, contained within the CD68+/FXIII-A- inflammatory pool. Despite it, in vitro, coculture experiments revealed that Ex-Ob ADSCs instructed monocyte differentiation into a M2-like profile, and under inflammatory conditions induced by LPS treatment, inhibited HLA-DR upregulation by resting M0 macrophages, originated a similar percentage of TNF-α+ cells, and inhibited IL-10 secretion, similar to OW-ADSCs and BMSCs, which were used for comparison, as these are the main alternative cell types available for therapeutic purposes. Our results showed that Ex-Ob ADSCs mirrored OW-ADSCs in macrophage education, favoring the M2 immunophenotype and a mixed (M1/M2) secretory response. These results have translational potential, since they provide evidence that ADSCs from both Ex-Ob and OW subjects can be used in regenerative medicine in eligible therapies. Further in vivo studies will be fundamental to validate these observations.
Collapse
Affiliation(s)
- Daiana V. Lopes Alves
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Integrated Laboratory of Morphology, Institute of Biodiversity and Sustainability, NUPEM, Federal University of Rio de Janeiro, Macaé, RJ, Brazil
| | - Cesar Claudio-da-Silva
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Surgery Department, Medical School, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcelo C. A. Souza
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Surgery Department, Medical School, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rosa T. Pinho
- Laboratory of Clinical Immunology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | | | - Radovan Borojevic
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carmen M. Nogueira
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hélio dos S. Dutra
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Christina M. Takiya
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Danielle C. Bonfim
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Maria Isabel D. Rossi
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
50
|
Ko E, Yoon T, Lee Y, Kim J, Park YB. ADSC secretome constrains NK cell activity by attenuating IL-2-mediated JAK-STAT and AKT signaling pathway via upregulation of CIS and DUSP4. Stem Cell Res Ther 2023; 14:329. [PMID: 37964351 PMCID: PMC10648656 DOI: 10.1186/s13287-023-03516-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: 05/01/2023] [Accepted: 09/25/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have immunomodulatory properties and therapeutic effects on autoimmune diseases through their secreted factors, referred to as the secretome. However, the specific key factors of the MSC secretome and their mechanisms of action in immune cells have not been fully determined. Most in vitro experiments are being performed using immune cells, but experiments using natural killer (NK) cells have been neglected, and a few studies using NK cells have shown discrepancies in results. NK cells are crucial elements of the immune system, and adjustment of their activity is essential for controlling various pathological conditions. The aim of this study was to elucidate the role of the adipose tissue-derived stem cell (ADSC) secretome on NK cell activity. METHODS To obtain the ADSC secretome, we cultured ADSCs in medium and concentrated the culture medium using tangential flow filtration (TFF) capsules. We assessed NK cell viability and proliferation using CCK-8 and CFSE assays, respectively. We analyzed the effects of the ADSC secretome on NK cell activity and pathway-related proteins using a combination of flow cytometry, ELISA, cytotoxicity assay, CD107a assay, western blotting, and quantitative real-time PCR. To identify the composition of the ADSC secretome, we performed LC-MS/MS profiling and bioinformatics analysis. To elucidate the molecular mechanisms involved, we used mRNA sequencing to profile the transcriptional expression of human blood NK cells. RESULTS The ADSC secretome was found to restrict IL-2-mediated effector function of NK cells while maintaining proliferative potency. This effect was achieved through the upregulation of the inhibitory receptor CD96, as well as downregulation of activating receptors and IL-2 receptor subunits IL-2Rα and IL-2Rγ. These changes were associated with attenuated JAK-STAT and AKT pathways in NK cells, which were achieved through the upregulation of cytokine-inducible SH2-containing protein (CIS, encoded by Cish) and dual specificity protein phosphatase 4 (DUSP4). Furthermore, proteomic analysis revealed twelve novel candidates associated with the immunomodulatory effects of MSCs. CONCLUSIONS Our findings reveal a detailed cellular outcome and regulatory mechanism of NK cell activity by the ADSC secretome and suggest a therapeutic tool for treating NK-mediated inflammatory and autoimmune diseases using the MSC secretome.
Collapse
Affiliation(s)
- Eunhee Ko
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Taejun Yoon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yoojin Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jongsun Kim
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|