1
|
Lu Y, Jin J, Zhang H, Lu Q, Zhang Y, Liu C, Liang Y, Tian S, Zhao Y, Fan H. Traumatic brain injury: Bridging pathophysiological insights and precision treatment strategies. Neural Regen Res 2026; 21:887-907. [PMID: 40145994 DOI: 10.4103/nrr.nrr-d-24-01398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 03/28/2025] Open
Abstract
Blood-brain barrier disruption and the neuroinflammatory response are significant pathological features that critically influence disease progression and treatment outcomes. This review systematically analyzes the current understanding of the bidirectional relationship between blood-brain barrier disruption and neuroinflammation in traumatic brain injury, along with emerging combination therapeutic strategies. Literature review indicates that blood-brain barrier disruption and neuroinflammatory responses are key pathological features following traumatic brain injury. In the acute phase after traumatic brain injury, the pathological characteristics include primary blood-brain barrier disruption and the activation of inflammatory cascades. In the subacute phase, the pathological features are characterized by repair mechanisms and inflammatory modulation. In the chronic phase, the pathological features show persistent low-grade inflammation and incomplete recovery of the blood-brain barrier. Various physiological changes, such as structural alterations of the blood-brain barrier, inflammatory cascades, and extracellular matrix remodeling, interact with each other and are influenced by genetic, age, sex, and environmental factors. The dynamic balance between blood-brain barrier permeability and neuroinflammation is regulated by hormones, particularly sex hormones and stress-related hormones. Additionally, the role of gastrointestinal hormones is receiving increasing attention. Current treatment strategies for traumatic brain injury include various methods such as conventional drug combinations, multimodality neuromonitoring, hyperbaric oxygen therapy, and non-invasive brain stimulation. Artificial intelligence also shows potential in treatment decision-making and personalized therapy. Emerging sequential combination strategies and precision medicine approaches can help improve treatment outcomes; however, challenges remain, such as inadequate research on the mechanisms of the chronic phase traumatic brain injury and difficulties with technology integration. Future research on traumatic brain injury should focus on personalized treatment strategies, the standardization of techniques, cost-effectiveness evaluations, and addressing the needs of patients with comorbidities. A multidisciplinary approach should be used to enhance treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Yujia Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Jie Jin
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Huajing Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Qianying Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Yingyi Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Chuanchuan Liu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Yangfan Liang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Sijia Tian
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Yanmei Zhao
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Haojun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| |
Collapse
|
2
|
Wang J, Luo Y, Wu Y, Du F, Shi S, Duan Y, Chen A, Zhang J, Yu S. Single-cell Raman spectroscopy as a novel platform for unveiling the heterogeneity of mesenchymal stem cells. Talanta 2025; 292:127933. [PMID: 40081243 DOI: 10.1016/j.talanta.2025.127933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/15/2025]
Abstract
Despite the significant potential of mesenchymal stem cells (MSC) therapy in clinical settings, challenges persist regarding the efficient detection of consistency and uniformity of MSC populations. Raman spectroscopy is a fast, convenient, and nondestructive technique to acquire molecular properties of biomolecules across laboratory and mass-production settings. Here we utilized Raman spectroscopy to evaluate the heterogeneity of primary MSC from varying donors, passages, and distinct culture conditions, and compared its effectiveness with conventional techniques such as flow cytometry. Although these MSC exhibited insignificant differences in morphology and surface markers in flow cytometry analysis, they could be distinctly clustered into different populations by Raman spectroscopy and the subsequent machine learning using linear discriminant analysis. Principal component analysis demonstrated limited efficiency in clustering Raman data from diverse sources, which could be enhanced through combination with support vector machine or deterministic finite automation. These findings highlight the sensitivity of Raman spectroscopy in detecting subtle differences. Moreover, the analysis of characteristic Raman peaks attributed to cellular biomolecules in MSC from passages 2 (P2) to P10 revealed a gradual decrease in the levels of nucleic acids, lipids, and proteins with increasing passages, and a significant increase in carotenoids from P8. These results suggest the potential use of Raman spectroscopy to assess cellular biochemical characteristics such as aging, with carotenoids emerging as a potential marker of cell aging. In conclusion, Raman spectroscopy demonstrates the ability to rapidly and non-invasively detect cellular heterogeneity and biochemical status, offering significant potential for quality control in stem cell therapy.
Collapse
Affiliation(s)
- Jingwen Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, 215163, China
| | - Yanjun Luo
- Shanghai D-Band Medical Technology Co., LTD, Shanghai, 201802, China
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, 215163, China
| | - Fangzhou Du
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, 215163, China
| | - Shuaiguang Shi
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yuhan Duan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Aoying Chen
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, 215163, China; Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China.
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, 215163, China; Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China.
| |
Collapse
|
3
|
Wijerathna-Yapa A, Isaac KS, Combe M, Hume S, Sokolenko S. Re-imagining human cell culture media: Challenges, innovations, and future directions. Biotechnol Adv 2025; 81:108564. [PMID: 40101881 DOI: 10.1016/j.biotechadv.2025.108564] [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: 12/16/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
The development of optimized culture media is pivotal to advancements in human cell culture, underpinning progress in regenerative medicine, cell therapies, and personalized medicine. While foundational formulations like Eagle's Minimum Essential Medium (MEM) and Dulbecco's Modified Eagle Medium (DMEM) have historically enabled significant biological research, these media were primarily designed for non-human cells and do not adequately address the unique metabolic and functional requirements of human cells. This review examines the evolution of cell culture media, identifying persistent challenges in reproducibility, scalability, and ethical concerns, particularly regarding the reliance on animal-derived components such as fetal bovine serum (FBS). We highlight innovations in serum-free and chemically defined media that offer promising alternatives by enhancing consistency, aligning with Good Manufacturing Practices, and addressing ethical concerns. Emerging approaches, including omics-based profiling, high-throughput screening, and artificial intelligence (AI)-driven media design, are reshaping media optimization by enabling precise tailoring to the needs of specific human cell types and patient-derived cells. Furthermore, we discuss economic and regulatory challenges, emphasizing the need for cost-effective and scalable solutions to facilitate clinical translation. Looking forward, integrating advanced biotechnological tools such as 3D bioprinting, organ-on-a-chip systems, and personalized media formulations presents a transformative opportunity for human cell culture. These innovations, aligned with ethical and clinical standards, can drive the development of human-specific media systems that ensure reproducibility, scalability, and enhanced therapeutic potential, thereby advancing both research and clinical applications.
Collapse
Affiliation(s)
- Akila Wijerathna-Yapa
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada
| | - Kathy Sharon Isaac
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada
| | - Michelle Combe
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada
| | - Samuel Hume
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Stanislav Sokolenko
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
4
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of stem cells in ageing and age-related diseases. Mech Ageing Dev 2025; 225:112069. [PMID: 40324541 DOI: 10.1016/j.mad.2025.112069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Stem cell functions and ageing are deeply interconnected, continually influencing each other in multiple ways. Stem cells play a vital role in organ maintenance, regeneration, and homeostasis, all of which decline over time due to gradual reduction in their self-renewal, differentiation, and growth factor secretion potential. The functional decline is attributed to damaging extrinsic environmental factors and progressively worsening intrinsic genetic and biochemical processes. These ageing-associated deteriorative changes have been extensively documented, paving the way for the discovery of novel biomarkers of ageing for detection, diagnosis, and treatment of age-related diseases. Age-dependent changes in adult stem cells include numerical decline, loss of heterogeneity, and reduced self-renewal and differentiation, leading to a drastic reduction in regenerative potential and thereby driving the ageing process. Conversely, ageing also adversely alters the stem cell niche, disrupting the molecular pathways underlying stem cell homing, self-renewal, differentiation, and growth factor secretion, all of which are critical for tissue repair and regeneration. A holistic understanding of these molecular mechanisms, through empirical research and clinical trials, is essential for designing targeted therapies to modulate ageing and improve health parameters in older individuals.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Global Research Alliances, Ashoka University, Rajiv Gandhi Education City, Sonepat, Haryana 131029, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
5
|
Li Z, Liu L, Sun Y, Liu X, Zhang P, Wang Y, Ding G. Mesenchymal stem/stromal cells-derived exosomes: possible therapeutic mechanism in inflammatory bowel disease. Hum Cell 2025; 38:111. [PMID: 40434563 DOI: 10.1007/s13577-025-01243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract caused by dysfunction of the immune system in genetically susceptible individuals. As current pharmacologic and surgical treatments remain suboptimal, increasing attention has been directed toward exosomes derived from mesenchymal stem/stromal cells (MSCs) as alternative therapeutic approaches. MSCs are multipotent stromal cells that can be isolated from various human tissues such as bone marrow, adipose, umbilical cord and periodontal ligament. Exosomes are cell-derived membrane-bound vesicles enclosing RNAs, proteins, growth factors, and cytokines. Previous studies indicate that the anti-inflammatory, immunomodulatory, and regenerative effects of MSCs are largely mediated by MSC-derived exosomes (MSC-Exos). Therefore, this review outlines current insights into the molecular mechanisms of MSC-Exos in IBD treatment to support the future development of MSC-Exos as a therapeutic strategy, thus providing novel observations into the clinical applications of MSC-Exos in IBD management.
Collapse
Affiliation(s)
- Zekun Li
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Luyun Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Yuhui Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Xinjuan Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Ping Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Yue Wang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China.
| |
Collapse
|
6
|
Pan M, Xu Y, Wang Y, Jiang Y, Xie Y, Tai C, Wang W, Wang B. The therapeutic efficacy comparison of MSCs derived different tissues unveilings anti-apoptosis more crucial than angiogenesis in treating acute myocardial infarction. Stem Cell Res Ther 2025; 16:236. [PMID: 40361236 PMCID: PMC12077008 DOI: 10.1186/s13287-025-04378-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Myocardial infarction (MI) is a severe disease that often associated with impaired angiogenesis and increased myocardial apoptosis. Mesenchymal stromal cells (MSCs) have been a promising candidate for treating myocardial infarction. However, functional heterogeneity of MSCs leads to inconsistent therapeutic efficiency and the current MSCs-based therapy lacks the concept and implementation of precision medicine. In this study, we compared the cardioprotective effect of UCMSCs and ADMSCs targeting the angiogenesis in a mouse MI model and screened out optimum MSCs candidate for precise clinical application. METHODS The gene expression profiles of UCMSCs and ADMSCs were investigated through RNA sequencing analysis. To compare their angiogenic potential, we performed tube formation assay, Matrigel plug assays, and aortic ring assay, and analyzed pro-angiogenic genes via qPCR. Subsequently, UCMSCs and ADMSCs were respectively injected into myocardium after MI surgery in mice. On day 28 post-MI, echocardiography was performed to assess cardiac function. Histological analysis was performed to assess MSCs retention, angiogenesis, and myocardial apoptosis. Additionally, the anti-apoptosis effects mediated by MSCs were further evaluated using flow cytometry in hypoxia H9C2 and HL-1 cells. RESULTS The RNA sequencing analysis revealed differences in gene expression related to angiogenesis and apoptosis pathways between UCMSCs and ADMSCs. UCMSCs presented greater pro-angiogenesis activity than ADMSCs in vitro and in vivo. Both of UCMSCs and ADMSCs improved cardiac function, decreased infarction area and inhibited cardiomyocyte apoptosis while promoting angiogenesis post-MI in mice. Notably, ADMSCs exerted a better cardioprotective function than UCMSCs and stronger anti-apoptotic effect on residual cardiomyocytes. CONCLUSIONS The protection of residual cells survival played a more prominent role than angiogenesis in MSCs-based therapy for acute MI. Our study provides new insights into therapeutic strategies and suggests that the optimal type of MSCs can be screened based on their tissue heterogeneity for precise clinical applications in acute MI.
Collapse
Affiliation(s)
- Mingjie Pan
- Clinical Medicine Research Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yueyue Xu
- The Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yaping Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital, Clinical Medical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yue Jiang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yuanyuan Xie
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Chenxu Tai
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Wenqing Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital, Clinical Medical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
| |
Collapse
|
7
|
Liu E, Sun Y, Yang L, Jiang H, Sun F, Chen L, Duan J, Yang S. Investigating the regulation of the miR-199a-3p/TGF-β/Smad signaling pathway by BSHXF drug-containing serum combined with ADSCs for delaying intervertebral disc degeneration. Front Pharmacol 2025; 16:1583635. [PMID: 40356987 PMCID: PMC12067415 DOI: 10.3389/fphar.2025.1583635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Background Intervertebral disc degeneration (IDD) significantly contributes to low back pain (LBP), yet effective treatment options are scarce. BSHXF, a classical traditional Chinese medicine formula, demonstrates dual pharmacological actions: tonifying kidneys, strengthening bones, activating blood circulation, and resolving stasis. It has been widely used in IDD management. Given its potential, combining BSHXF with miRNA regulation and stem cell therapy may enhance therapeutic outcomes by targeting molecular and cellular pathways underlying IDD pathogenesis. Aim of the study IDD is recognized as one of the primary causes of low back pain, yet effective therapeutic interventions for this condition remain limited. This study explores the role of BSHXF drug-containing serum combined with adipose-derived stem cells (ADSCs) in slowing IDD progression via the miR-199a-3p/TGF-β/Smad signaling pathway. By comprehensively investigating the synergistic effects of this combination therapy, we aim to propose a novel multi-target strategy that addresses the complex pathogenesis of IDD. Materials and Methods This study employed a combination of in vivo and in vitro models. An IDD model was induced in rat caudal intervertebral discs through needle puncture, while an oxidative stress-induced ADSCs injury model was created in vitro using tert-butyl hydroperoxide (T-BHP). Cell viability was measured with the CCK-8 assay. Cell cycle distribution and mitochondrial reactive oxygen species (ROS) levels were assessed using flow cytometry. Cellular senescence was assessed using SA-β-galactosidase staining. Lactate dehydrogenase (LDH) activity was quantified to evaluate cellular damage. Differentiation into nucleus pulposus-like cells was assessed using immunofluorescence double staining for CD73 and COL2A1. ELISA was used to measure inflammatory cytokines (TNF-α, IL-1β, IL-4, IL-10) in cell supernatants. miR-199a-3p expression was determined using RT-qPCR. Western blotting was employed to quantify COL2A1, SOX9, and ACAN protein levels, reflecting nucleus pulposus-like differentiation and extracellular matrix (ECM) synthesis capacity. Western blotting was employed to assess pathway activity by analyzing the protein expressions of TGF-β1, Smad2, Smad3, and their phosphorylated forms, P-Smad2 and P-Smad3. In vivo experiments assessed histopathological degeneration through hematoxylin-eosin (HE) and Safranin O-Fast Green staining. Immunohistochemistry (IHC) analyzed COL1A1 and COL2A1 expression levels. RT-qPCR quantified miR-199a-3p expression. Western blotting was employed to assess the expression levels of TGF-β1, Smad2, Smad3, P-Smad2, and P-Smad3 for pathway regulation evaluation. Results Our experimental results demonstrated that serum containing BSHXF significantly alleviated T-BHP-induced oxidative stress, improved the cellular microenvironment, promoted ADSCs proliferation, and decelerated cellular senescence. Further mechanistic analysis revealed that BSHXF significantly activated the TGF-β/Smad signaling pathway, driving the differentiation of ADSCs into nucleus pulposus-like cells and restoring normal cell cycle progression. Overexpression of miR-199a-3p inhibited the TGF-β/Smad pathway, leading to ECM degradation and elevated expression of inflammatory factors (TNF-α, IL-1β). In contrast, BSHXF restored TGF-β/Smad pathway activity by downregulating miR-199a-3p expression. In vivo experiments demonstrated that miR-199a-3p overexpression exacerbated IDD, characterized by reduced COL2A1 expression, elevated COL1A1 levels, and increased disc fibrosis. BSHXF intervention markedly attenuated IDD progression by downregulating miR-199a-3p expression, reducing disc fibrosis, and effectively restoring collagen expression. Conclusion BSHXF activated the TGF-β/Smad pathway to promote the differentiation of ADSCs into nucleus pulposus-like cells. It exerted protective effects by alleviating oxidative stress damage, improving the microenvironment, delaying senescence, and enhancing cellular functions. This study is the first to reveal that miR-199a-3p overexpression exacerbates intervertebral disc fibrosis and degeneration. BSHXF restored TGF-β/Smad pathway activity by downregulating miR-199a-3p expression, thereby improving disc structure and function. This integrated approach offers a novel multi-target intervention strategy for IDD, demonstrating significant therapeutic potential.
Collapse
Affiliation(s)
- Enxu Liu
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| | - Yu Sun
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| | - Lei Yang
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| | - Haobo Jiang
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| | - Fei Sun
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
| | - Long Chen
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| | - Jiahao Duan
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| | - Shaofeng Yang
- Hunan University of Traditional Chinese Medicine, Graduate School, Changsha, Hunan, China
- The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Department of Orthopaedics, Changsha, Hunan, China
| |
Collapse
|
8
|
Giler MK, Tucker HA, Foote AK, Francis AG, Madsen SD, Liu YZ, O’Connor KC. RNA sequencing identifies MAP1A and PTTG1 as predictive genes of aging CD264 + human mesenchymal stem cells at an early passage. Cytotechnology 2025; 77:63. [PMID: 39980838 PMCID: PMC11839963 DOI: 10.1007/s10616-025-00724-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Molecular profiles of mesenchymal stem cells (MSCs) are needed to standardize the composition and effectiveness of MSC therapeutics. This study employs RNA sequencing to identify genes to be used in concert with CD264 as a molecular profile of aging MSCs at a clinically relevant culture passage. CD264- and CD264+ populations were isolated by fluorescence-activated cell sorting from passage 4 MSC cultures. CD264+ MSCs exhibited an aging phenotype relative to their CD264- counterpart. Donor-matched CD264-/+ mRNA samples from 5 donors were subjected to pair-ended, next-generation sequencing. An independent set of 5 donor MSCs was used to validate differential expression of select genes with quantitative reverse transcription PCR. Pairwise differential expression analysis identified 2,322 downregulated genes and 2,695 upregulated genes in CD264+ MSCs relative to donor-matched CD264- MSCs with a Benjamini-Hochberg adjusted p-value (BH p adj ) < 0.1. Nearly 25% of these genes were unique to CD264-/+ MSCs and not differentially expressed at a significance level of BH p adj < 0.1 in previous RNA sequencing studies of early- vs. late-passage MSCs. Least Absolute Shrinkage and Selection Operator regression identified microtubule-associated protein 1A (MAP1A) and pituitary tumor-transforming gene 1 (PTTG1) as predictive genes of CD264+ MSCs. Combined MAP1A and PTTG1 expression correctly classified CD264 status of MSC samples with an accuracy of 100%. Differential expression and predictive ability of MAP1A and PTTG1 compared favorably with that of existing senescence markers expressed in early passage CD264-/+ MSCs. This study provides the first linkage of MAP1A to CD264, aging and senescence. Our findings have application as quality metrics to standardize the composition of MSC therapies and as molecular targets to slow/reverse cellular aging. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00724-8.
Collapse
Affiliation(s)
- Margaret K. Giler
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA USA
| | - H. Alan Tucker
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA USA
| | - Amanda K. Foote
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA USA
| | - Avery G. Francis
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA USA
| | - Sean D. Madsen
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA USA
| | - Yao-Zhong Liu
- Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA USA
| | - Kim C. O’Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA USA
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA USA
- Center for Aging, School of Medicine, Tulane University, New Orleans, LA USA
| |
Collapse
|
9
|
Liang L, Liu H, Wang S. Placental mesenchymal stem cell-derived exosomes treat endometrial injury in a rat model of intrauterine adhesions. Mol Genet Genomics 2025; 300:36. [PMID: 40128387 PMCID: PMC11933197 DOI: 10.1007/s00438-025-02241-x] [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: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/26/2025]
Abstract
Intrauterine adhesion (IUA) refer to persistent inflammation and fibrosis due to damaged or infected endometrium and eventually lead to dysfunction. This study aimed to explore the therapeutic effects of exosomes (Exos) derived from placental mesenchymal stem cells (PMSCs) on endometrial repair in a rat model of IUA and to elucidate the underlying molecular mechanisms. PMSCs were characterized using flow cytometry and differentiation assays (osteogenic, adipogenic, and chondrogenic). Exos were isolated via ultracentrifugation and validated through transmission electron microscopy, nanoparticle tracking analysis and Western blot. An IUA model was established via electrocoagulation, and endometrial repair was assessed using hematoxylin-eosin (HE) and Masson staining. RNA sequencing, differential expression analysis and protein-protein interaction (PPI) network construction were employed to investigate the molecular mechanisms of PMSC Exos mediated repair. The role of miR-143 in targeting MyD88 and modulating the NF-κB signaling pathway was confirmed using Dual-Luciferase Reporter Assay and qRT-PCR. PMSC Exos significantly improved endometrial thickness, increased glandular number and reduced fibrosis in the IUA model. RNA sequencing and differential expression analysis screened 3980 differentially expressed genes (DEGs) common to the IUA vs normal groups and Exo vs IUA groups. Enrichment analysis revealed significant involvement of immune system processes, natural killer cell-mediated cytotoxicity and NF-κB signaling. PMSC Exos delivered miR-143, which targeted MyD88, thereby regulating the NF-κB pathway. PMSC Exos effectively repaired endometrial damage in the IUA model by modulating the NF-κB signaling pathway through miR-143 delivery. These findings suggest that PMSC Exos hold promise as a novel therapeutic strategy for IUA, offering insights into the molecular mechanisms underlying endometrial repair.
Collapse
Affiliation(s)
- Lin Liang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1, Dahuaroad, Dongdan, Beijing, 100730, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Dong Dan Santiao, Beijing, 100730, P.R. China
| | - Huidong Liu
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1, Dahuaroad, Dongdan, Beijing, 100730, P.R. China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Dong Dan Santiao, Beijing, 100730, P.R. China
| | - Shaowei Wang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1, Dahuaroad, Dongdan, Beijing, 100730, P.R. China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Graduate School of Peking Union Medical College, Dong Dan Santiao, Beijing, 100730, P.R. China.
| |
Collapse
|
10
|
Giebel B. A milestone for the therapeutic EV field: FDA approves Ryoncil, an allogeneic bone marrow-derived mesenchymal stromal cell therapy. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:183-190. [PMID: 40206802 PMCID: PMC11977348 DOI: 10.20517/evcna.2025.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025]
Abstract
Small extracellular vesicles (sEVs) derived from mesenchymal stromal cells (MSCs) hold substantial promise for therapeutic applications, including immune modulation and tissue regeneration. However, challenges such as batch-to-batch variability, donor material diversity, and the lack of standardized potency testing remain significant barriers to clinical translation. The recent U.S. Food and Drug Administration (FDA) approval of Ryoncil (remestemcel-L) for steroid-refractory acute graft-versus-host disease (aGvHD) in pediatric patients represents a crucial milestone for MSC-based therapies, offering also valuable insights for the development of MSC-EV therapies. This approval highlights the critical need to address variability and standardization issues in MSC products. Strategies like immortalizing MSCs and expanding them clonally can improve scalability, consistency, and overcome limitations inherent to cellular MSC therapies. With the FDA's decision signaling significant progress, optimizing MSC expansion protocols and refining potency testing methods will be crucial for advancing MSC-EVs as a viable therapeutic option, overcoming current challenges, and expanding clinical applications.
Collapse
Affiliation(s)
- Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| |
Collapse
|
11
|
Wei X, Mu H, Zhang Q, Zhang Z, Ru Y, Lai K, Ma Y, Lin Z, Tuxun R, Chen Z, Xiang AP, Li T. MSCs act as biopatches for blood-retinal barrier preservation to enhance functional recovery after retinal I/R. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102445. [PMID: 39967853 PMCID: PMC11834101 DOI: 10.1016/j.omtn.2024.102445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025]
Abstract
Retinal ischemia/reperfusion (I/R) is one of the most common pathologies of many vision-threatening diseases and is caused by blood-retinal barrier (BRB) breakdown and the resulting inflammatory infiltration. Targeting BRB is promising for retinal I/R treatment. Mesenchymal stromal cells (MSCs) are emerging as novel therapeutic strategies. Although intravitreal injection targets the retina, the restricted number of injected cells still requires the precise biodistribution of MSCs near the injury site. Here, we found that retinal I/R led to BRB breakdown, which induced protein and cell leakage from the circulation. Retinal cell death and diminished visual function were subsequently detected. Moreover, the expression of the chemokine CCL5 increased after retinal I/R, and CCL5 colocalized with the BRB. We then overexpressed CCR5 in human induced pluripotent stem cell-derived MSCs (iMSCs). In vivo, intravitreal-injected iMSCCCR5 preferentially migrated and directly integrated into the BRB, which preferably restored BRB integrity and eventually promoted retinal function recovery after retinal I/R. In summary, our work suggested that iMSCs act as biopatches for BRB preservation and that iMSC-based therapy is a promising therapeutic approach for retinal diseases related to I/R.
Collapse
Affiliation(s)
- Xiaoyue Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| | - Hanyiqi Mu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qinmu Zhang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ziyuan Zhang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yifei Ru
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Kunbei Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| | - Yuan Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| | - Zhuangling Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| | - Rebiya Tuxun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| | - Zitong Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tao Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, China
| |
Collapse
|
12
|
Bonfield TL, Lazarus HM. Human mesenchymal stem cell therapy: Potential advances for reducing cystic fibrosis infection and organ inflammation. Best Pract Res Clin Haematol 2025; 38:101602. [PMID: 40274338 DOI: 10.1016/j.beha.2025.101602] [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: 11/20/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Innovation in cystic fibrosis (CF) supportive care, including implementing new antimicrobial agents, improved physiotherapy, and highly effective modulators therapy, has advanced patient survival into the 4th and 5th decades of life. However, even with these remarkable improvements in therapy, CF patients continue to suffer from pulmonary infection and other visceral organ complications associated with long-term deficient cystic fibrosis transmembrane conductance regulator (CFTR) expression. Human mesenchymal stem cells (MSCs) have been utilized in tissue engineering based upon their capacity to provide structural components of mesenchymal tissues. An alternative role of MSCs, however is their versatile utilization as cell-based infusion powerhouses due to the unique capacity to deliver milieu specific soluble biologic factors, promoting immune supportive antimicrobial and anti-inflammatory potency. MSCs derived from umbilical cord blood, bone marrow, adipose and other tissues can be expanded in ex vivo using good manufacturing procedure facilities for a safe, unique therapeutic to reduce and limit CF infection and facilitate the resolution of multi-organ inflammation. In our efforts, we conducted extensive preclinical development and validation of an allogeneic derived bone marrow derived MSC product in preparation for a clinical trial in CF. In this process, potency models were developed to ensure the functional capacity of the MSC product to provide clinical benefit. In vitro, murine in vivo and patient tissue ex vivo potency models were utilized to follow MSC anti-infective and anti-inflammatory potency associated with the CFTR deficient environment. We showed in our "First in CF" clinical trial that the allogeneic MSCs obtained from healthy volunteer bone marrow samples were safe. The advent of improved CF care measures and exciting new small molecules has changed the survival and morbidity phenotype of patients with CF, however, there are CF patients who cannot tolerate or have genotypes that are non-responsive to modulators. Additionally, even with the small molecule therapy, CF patients are living longer, but without genetic correction, with the CF disease manifestation aggravated by the continuance of pre-existing CFTR-associated clinical issues such as ongoing inflammation. MSCs secrete bio-active factors that enhance and protect tissue function and can promote "self-immune" regulation. These properties can provide therapeutic support for the traditional and changing face of CF disease clinical complications. Further, MSC-derived bio-active factors can directly mitigate colonizing pathogens' survival by producing antimicrobial peptides (AMPs) which change the pathogen surface and increase host recognition, elimination, and sensitivity to antibiotics. Herein, we review the potential of MSC therapeutics for treating many facets of CF, emphasizing the potential for providing great additive therapeutics for managing morbidity and quality of life.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Genetics and Genome Sciences, National Center for Regenerative Medicine, Pediatrics and Pathology, Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| | - Hillard M Lazarus
- Department of Medicine, Hematology and National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
13
|
Watanabe M, Asawa Y, Riu D, Sakamoto T, Hoshi K, Hikita A. Identification of mesenchymal stem cell populations with high osteogenic potential using difference in cell division rate. Regen Ther 2025; 28:498-508. [PMID: 39991510 PMCID: PMC11846930 DOI: 10.1016/j.reth.2025.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction In bone regenerative medicine, mesenchymal stem cells (MSCs) have been widely investigated for their potential in bone regeneration. However, MSCs are a heterogeneous cell population containing a variety of cell types, making it difficult to obtain a homogeneous MSC population sufficient for tissue regeneration. Our group previously reported that by selecting rapidly dividing human auricular chondrocytes, it was possible to enrich for more chondrogenic cells. In this study, we aimed to identify a highly osteogenic MSC population by using a similar approach for mouse bone marrow MSCs. Methods Mouse bone marrow MSCs were fluorescently labeled with carboxyfluorescein succinimidyl ester (CFSE) and sorted according to the fluorescence intensity using flow cytometry on day 3 after labeling. To compare the ability to produce bone matrix in vitro, osteogenic differentiation cultures were performed and mineral deposition was confirmed by alizarin red staining. Real-time qPCR was also performed to examine the differences in gene expression between the fast- and slow-dividing cell groups immediately after aliquoting and after osteogenic differentiation. Results Differences in the growth rate of the fractionated cells were maintained after culture. Results of osteogenic differentiation culture and alizarin red staining showed more extensive mineral deposition in the slow cell group than in the fast cell group. Calcium quantification also showed higher absorbance in the slow cell group compared to the fast cell group, indicating higher osteogenic differentiation potential in the slow cell group. Furthermore, real-time qPCR analysis showed that osteocalcin expression was higher in the slow cell group in cells immediately after preparative differentiation. In addition, the expression of osteocalcin and sclerostin were higher in the slow cells after osteogenic differentiation. Conclusion The slow cell population contains many highly differentiated cells that are already more deeply committed to the bone lineage, suggesting that they have higher osteogenic differentiation potential than the fast cell population. This study will contribute to the realization of better bone regenerative medicine by utilizing the high osteogenic differentiation potential of the slow cell population.
Collapse
Affiliation(s)
- Maya Watanabe
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yukiyo Asawa
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Dan Riu
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoaki Sakamoto
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Oral-maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Atsuhiko Hikita
- Division of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
14
|
Revokatova D, Koteneva P, Kosheleva N, Shpichka A, Timashev P. Spheroids from Epithelial and Mesenchymal Cell Phenotypes as Building Blocks in Bioprinting (Review). Sovrem Tekhnologii Med 2025; 17:133-154. [PMID: 40071071 PMCID: PMC11892564 DOI: 10.17691/stm2025.17.1.11] [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: 11/19/2024] [Indexed: 03/14/2025] Open
Abstract
Most tissues and organs are based on cells of the epithelial and mesenchymal phenotypes. Epithelial cells build protective barriers, have a key role in absorption and secretion, and participate in metabolism. Characterized by high plasticity and ability to migrate, mesenchymal cells ensure structural support, promote tissue restoration and are important for matrix remodeling. Interaction between these two cell types is critical for maintaining the body integrity and functioning. Modern tissue engineering is aimed at creation of artificial tissues and organs that have the required cellular composition, mechanical properties and functional potential for medical usage. One of the most popular methods of tissue engineering is 3D bioprinting, which allows creating complex three-dimensional structures with specified characteristics. Recently, special attention has been paid to bioprinting with spheroids being three-dimensional cellular aggregates that can be used as building blocks for tissue-engineered structures. Due to numerous cell-to-cell contacts and accumulation of extracellular matrix, spheroids ensure conditions allowing to form anatomical tissues and organs. To optimize bioprinting conditions, one shall precisely understand the mechanical properties of spheroids, as they directly affect the ability of cells to migrate and fuse, and thus the rate of construct formation and its overall morphology. This review summarizes the available data on the differences in mechanical properties of epithelial and mesenchymal spheroids, examines methods for their co-culturing in various applications of regenerative medicine, as well as analyzes the peculiarities of their use in different bioprinting methods to obtain high-quality tissue constructs.
Collapse
Affiliation(s)
- D.P. Revokatova
- Junior Researcher, Laboratory of Clinical Smart- and Nanotechnologies, Institute of Regenerative Medicine; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| | - P.I. Koteneva
- Junior Researcher, Biofabrika Design Center, Institute of Regenerative Medicine; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| | - N.V. Kosheleva
- PhD, Associate Professor, Head of Laboratory of Clinical Smart- and Nanotechnologies, Institute of Regenerative Medicine; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| | - A.I. Shpichka
- PhD, Associate Professor, Head of Laboratory of Applied Microfluidics, Institute of Regenerative Medicine; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| | - P.S. Timashev
- DSc, Professor, Institute of Regenerative Medicine; Chief Scientific Officer of the Scientific and Technological Park of Biomedicine; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| |
Collapse
|
15
|
Goss K, Horwitz EM. Single-cell multiomics to advance cell therapy. Cytotherapy 2025; 27:137-145. [PMID: 39530970 DOI: 10.1016/j.jcyt.2024.10.009] [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: 09/13/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Single-cell RNA-sequencing (scRNAseq) was first introduced in 2009 and has evolved with many technological advancements over the last decade. Not only are there several scRNAseq platforms differing in many aspects, but there are also a large number of computational pipelines available for downstream analyses which are being developed at an exponential rate. Such computational data appear in many scientific publications in virtually every field of study; thus, investigators should be able to understand and interpret data in this rapidly evolving field. Here, we discuss key differences in scRNAseq platforms, crucial steps in scRNAseq experiments, standard downstream analyses and introduce newly developed multimodal approaches. We then discuss how single-cell omics has been applied to advance the field of cell therapy.
Collapse
Affiliation(s)
- Kyndal Goss
- Marcus Center for Advanced Cellular Therapy, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Graduate Division of Biology and Biomedical Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA
| | - Edwin M Horwitz
- Marcus Center for Advanced Cellular Therapy, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Graduate Division of Biology and Biomedical Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA.
| |
Collapse
|
16
|
Wan R, Liu Y, Yan J, Lin J. Cell therapy: A beacon of hope in the battle against pulmonary fibrosis. FASEB J 2025; 39:e70356. [PMID: 39873972 DOI: 10.1096/fj.202402790r] [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: 11/10/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterized by abnormal activation of myofibroblasts and pathological remodeling of the extracellular matrix, with a poor prognosis and limited treatment options. Lung transplantation is currently the only approach that can extend the life expectancy of patients; however, its applicability is severely restricted due to donor shortages and patient-specific limitations. Therefore, the search for novel therapeutic strategies is imperative. In recent years, stem cells have shown great promise in the field of regenerative medicine due to their self-renewal capacity and multidirectional differentiation potential, and a growing body of literature supports the efficacy of stem cell therapy in PF treatment. This paper systematically summarizes the research progress of various stem cell types in the treatment of PF. Furthermore, it discusses the primary methods and clinical outcomes of stem cell therapy in PF, based on both preclinical and clinical data. Finally, the current challenges and key factors to consider in stem cell therapy for PF are objectively analyzed, and future directions for improving this therapy are proposed, providing new insights and references for the clinical treatment of PF patients.
Collapse
Affiliation(s)
- Ruyan Wan
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jingwen Yan
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
17
|
da Silva KN, Marim FM, Rocha GV, Costa-Ferro ZSM, França LSDA, Nonaka CKV, Paredes BD, Rossi EA, Loiola EC, Adanho CSA, Cunha RS, Silva MMAD, Cruz FF, Costa VV, Zanette DL, Rocha CAG, Aguiar RS, Rocco PRM, Souza BSDF. Functional heterogeneity of mesenchymal stem cells and their therapeutic potential in the K18-hACE2 mouse model of SARS-CoV-2 infection. Stem Cell Res Ther 2025; 16:15. [PMID: 39849557 PMCID: PMC11756204 DOI: 10.1186/s13287-024-04086-4] [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: 05/07/2024] [Accepted: 11/28/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Despite many years of investigation into mesenchymal stem cells (MSCs) and their potential for treating inflammatory conditions such as COVID-19, clinical outcomes remain variable due to factors like donor variability, different tissue sources, and diversity within MSC populations. Variations in MSCs' secretory and proliferation profiles, and their proteomic and transcriptional characteristics significantly influence their therapeutic potency, highlighting the need for enhanced characterization methods to better predict their efficacy. This study aimed to evaluate the biological characteristics of MSCs from different tissue origins, selecting the most promising line for further validation in a K18-hACE2 mouse model of SARS-CoV-2 infection. METHODS We studied nine MSC lines sourced from either bone marrow (hBMMSC), dental pulp (hDPMSC), or umbilical cord tissue (hUCMSC). The cells were assessed for their proliferative capacity, immunophenotype, trilineage differentiation, proteomic profile, and in vitro immunomodulatory potential by co-culture with activated lymphocytes. The most promising MSC line was selected for further experimental validation using the K18-hACE2 mouse model of SARS-CoV-2 infection. RESULTS The analyzed cells met the minimum criteria for defining MSCs, including the expression of surface molecules and differentiation capacity, showing genetic stability and proliferative potential. Proteomic analysis revealed distinct protein profiles that correlate with the tissue origin of MSCs. The immunomodulatory response exhibited variability, lacking a discernible pattern associated with their origin. In co-culture assays with lymphocytes activated with anti-CD3/CD28 beads, all MSC lines demonstrated the ability to inhibit TNF-α, to induce TGF-β and Indoleamine 2,3-dioxygenase (IDO), with varying degrees of inhibition observed for IFN-γ and IL-6, or induction of IL-10 expression. A module of proteins was found to statistically correlate with the potency of IL-6 modulation, leading to the selection of one of the hUCMSCs as the most promising line. Administration of hUCMSC to SARS-CoV-2-infected K18 mice expressing hACE2 was effective in improving lung histology and modulating of a panel of cytokines. CONCLUSIONS Our study assessed MSCs derived from various tissues, uncovering significant variability in their characteristics and immunomodulatory capacities. Particularly, hUCMSCs demonstrated potential in mitigating lung pathology in a SARS-CoV-2 infection model, suggesting their promising therapeutic efficacy.
Collapse
Affiliation(s)
- Kátia Nunes da Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gisele Vieira Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | | | | | | | | | - Erik Aranha Rossi
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Erick Correia Loiola
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | | | - Rachel Santana Cunha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Mayck Medeiros Amaral da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Clarissa Araújo Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Renato Santana Aguiar
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil.
| |
Collapse
|
18
|
Li X, Cui J, Ning LJ, Hu RN, Zhao LL, Luo JJ, Xie XY, Zhang YJ, Luo JC, Li ZY, Qin TW. Response of a tenomodulin-positive subpopulation of human adipose-derived stem cells to decellularized tendon slices. Biomed Mater 2025; 20:025004. [PMID: 39746323 DOI: 10.1088/1748-605x/ada509] [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: 10/22/2024] [Accepted: 01/02/2025] [Indexed: 01/04/2025]
Abstract
The selection of appropriate cell sources is vital for the regeneration and repair of tendons using stem cell-based approaches. Human adipose-derived stem cells (hADSCs) have emerged as a promising therapeutic strategy for tendon injuries. However, the heterogeneity of hADSCs can lead to inconsistent or suboptimal therapeutic outcomes. In this study, we isolated and identified a tenomodulin (TNMD)-positive subpopulation from hADSCs (TNMD+hADSCs) using flow cytometry and then assessed the cellular response of this subpopulation to decellularized tendon slices (DTSs), including cell proliferation, migration, and tenogenic differentiation, using the CCK-8 assay, transwell migration assay, and quantitative real-time polymerase chain reaction. Our findings revealed that TNMD+hADSCs maintained the general characteristics of stem cells and exhibited significantly higher expressions of tendon-related markers compared to hADSCs. Importantly, DTSs significantly enhanced the proliferation, migration, and tenogenic differentiation of TNMD+hADSCs. This study provides preliminary experimental evidence for the translational application of ADSCs for tendon regeneration and repair.
Collapse
Affiliation(s)
- Xuan Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jing Cui
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Liang-Ju Ning
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ruo-Nan Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lei-Lei Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jia-Jiao Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xin-Yue Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yan-Jing Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jing-Cong Luo
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zheng-Yong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ting-Wu Qin
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cells and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
19
|
Beetler DJ, Giresi P, Di Florio DN, Fliess JJ, McCabe EJ, Watkins MM, Xu V, Auda ME, Bruno KA, Whelan ER, Kocsis SPC, Edenfield BH, Walker S, Macomb LP, Keegan KC, Jain A, Morales-Lara AC, Chekuri I, Hill AR, Farres H, Wolfram J, Behfar A, Stalboerger PG, Terzic A, Cooper L, Fairweather D. Therapeutic effects of platelet-derived extracellular vesicles on viral myocarditis correlate with biomolecular content. Front Immunol 2025; 15:1468969. [PMID: 39835120 PMCID: PMC11743460 DOI: 10.3389/fimmu.2024.1468969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Extracellular vesicles (EVs) can potently inhibit inflammation yet there is a lack of understanding about the impact of donor characteristics on the efficacy of EVs. The goal of this study was to determine whether the sex and age of donor platelet-derived EVs (PEV) affected their ability to inhibit viral myocarditis. Methods PEV, isolated from men and women of all ages, was compared to PEV obtained from women under 50 years of age, which we termed premenopausal PEV (pmPEV). Because of the protective effect of estrogen against myocardial inflammation, we hypothesized that pmPEV would be more effective than PEV at inhibiting myocarditis. We injected PEV, pmPEV, or vehicle control in a mouse model of viral myocarditis and examined histology, gene expression, protein profiles, and performed proteome and microRNA (miR) sequencing of EVs. Results We found that both PEV and pmPEV significantly inhibited myocarditis; however, PEV was more effective, which was confirmed by a greater reduction of inflammatory cells and proinflammatory and profibrotic markers determined using gene expression and immunohistochemistry. Proteome and miR sequencing of EVs revealed that PEV miRs specifically targeted antiviral, Toll-like receptor (TLR)4, and inflammasome pathways known to contribute to myocarditis while pmPEV contained general immunoregulatory miRs. Discussion These differences in EV content corresponded to the differing anti-inflammatory effects of the two types of EVs on viral myocarditis.
Collapse
Affiliation(s)
- Danielle J. Beetler
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Presley Giresi
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Damian N. Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Jessica J. Fliess
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Elizabeth J. McCabe
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Molly M. Watkins
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Vivian Xu
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Matthew E. Auda
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, United States
| | - Emily R. Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Stephen P. C. Kocsis
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | | | - Sierra A. Walker
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Logan P. Macomb
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Kevin C. Keegan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Angita Jain
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | | | - Isha Chekuri
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Anneliese R. Hill
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Houssam Farres
- Department of Vascular Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Joy Wolfram
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Atta Behfar
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, United States
| | - Paul G. Stalboerger
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, United States
| | - Andre Terzic
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, United States
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, United States
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
20
|
Kawatani K, Omana Suarez G, Perkerson RB, Parent EE, Nambara T, Knight JA, Parsons TM, Gupta K, Shue F, Alnobani A, Vibhute P, Cai H, Guerrero-Cázares H, Copland JA, Quiñones-Hinojosa A, Kanekiyo T. Human iPSC-Derived MSCs Induce Neurotrophic Effects and Improve Metabolic Activity in Acute Neuronal Injury Models. J Neurosci 2025; 45:e0606242024. [PMID: 39496487 DOI: 10.1523/jneurosci.0606-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/15/2024] [Accepted: 10/25/2024] [Indexed: 11/06/2024] Open
Abstract
Mesenchymal stromal cell (MSC) therapy has regenerative potentials to treat various pathological conditions including neurological diseases. MSCs isolated from various organs can differentiate into specific cell types to repair organ damages. However, their paracrine mechanisms are predicted to predominantly mediate their immunomodulatory, proangiogenic, and regenerative properties. While preclinical studies highlight the significant potential of MSC therapy in mitigating neurological damage from stroke and traumatic brain injury, the variability in clinical trial outcomes may stem from the inherent heterogeneity of somatic MSCs. Accumulating evidence has demonstrated that induced pluripotent stem cells (iPSCs) are an ideal alternative resource for the unlimited expansion and biomanufacturing of MSCs. Thus, we investigated how iPSC-derived MSCs (iMSCs) influence properties of iPSC-derived neurons. Our findings demonstrate that the secretome from iMSCs possesses neurotrophic effects, improving neuronal survival and promoting neuronal outgrowth and synaptic activity in vitro. Additionally, the iMSCs enhance metabolic activity via mitochondrial respiration in neurons, both in vitro and in mouse models. Glycolytic pathways also increased following the administration of iMSC secretome to iPSC-derived neurons. Consistently, in vivo experiments showed that intravenous administration of iMSCs compensated for the elevated energetic demand in male mice with irradiation-induced brain injury by restoring synaptic metabolic activity during acute brain damage. 18F-FDG PET imaging also detected an increase in brain glucose uptake following iMSC administration. Together, our results highlight the potential of iMSC-based therapy in treating neuronal damage in various neurological disorders, while paving the way for future research and potential clinical applications of iMSCs in regenerative medicine.
Collapse
Affiliation(s)
- Keiji Kawatani
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Genesis Omana Suarez
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, Florida 32224
| | - Ralph B Perkerson
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida 32224
| | - Ephraim E Parent
- Departments of Radiology, Mayo Clinic, Jacksonville, Florida 32224
| | - Toshihiko Nambara
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | | | - Tammee M Parsons
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida 32224
| | - Kshama Gupta
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Alla Alnobani
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Prasanna Vibhute
- Departments of Radiology, Mayo Clinic, Jacksonville, Florida 32224
| | - Hancheng Cai
- Departments of Radiology, Mayo Clinic, Jacksonville, Florida 32224
| | - Hugo Guerrero-Cázares
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
- Neurosurgery, Mayo Clinic, Jacksonville, Florida 32224
| | - John A Copland
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
| | - Alfredo Quiñones-Hinojosa
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Cancer Biology, Mayo Clinic, Jacksonville, Florida 32224
- Neurosurgery, Mayo Clinic, Jacksonville, Florida 32224
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, Florida 32224
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida 32224
| |
Collapse
|
21
|
Rasouli M, Alavi M, D'Angelo A, Sobhani N, Roudi R, Safari F. Exploring the dichotomy of the mesenchymal stem cell secretome: Implications for tumor modulation via cell-signaling pathways. Int Immunopharmacol 2024; 143:113265. [PMID: 39353385 DOI: 10.1016/j.intimp.2024.113265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Current cancer therapeutic strategies for the treatment of cancer are often unsuccessful due to unwanted side effects and drug resistance. Therefore, the design and development of potent, new anticancer platforms, such as stem-cell treatments, have attracted much attention. Distinctive biological properties of stem cells include their capacity to secrete bioactive factors, their limited immunogenicity, and their capacity for renewing themselves. Mesenchymal stem cells (MSCs) are one of several kinds of stem cells that are conveniently extracted and are able to be cultivated in vitro utilizing various sources. The secretome of stem cells contains many trophic factors, including cytokines, chemokines, growth factors, and microRNA molecules that can either promote or inhibit the formation of tumors, based on the cell environment. In the current review, we focused on the secretome of mesenchymal stem cells. These stem cells act as a double-edged sword in the regulation of cell signal transduction pathways in that they can either suppress or promote tumors.
Collapse
Affiliation(s)
- Mohammad Rasouli
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mana Alavi
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Alberto D'Angelo
- Oncology Department, Royal United Hospital, Bath BA1 3NG, United Kingdom
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA.
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| |
Collapse
|
22
|
İyisan N, Hausdörfer O, Wang C, Hiendlmeier L, Harder P, Wolfrum B, Özkale B. Mechanoactivation of Single Stem Cells in Microgels Using a 3D-Printed Stimulation Device. SMALL METHODS 2024; 8:e2400272. [PMID: 39011729 PMCID: PMC11672187 DOI: 10.1002/smtd.202400272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/03/2024] [Indexed: 07/17/2024]
Abstract
In this study, the novel 3D-printed pressure chamber for encapsulated single-cell stimulation (3D-PRESS) platform is introduced for the mechanical stimulation of single stem cells in 3D microgels. The custom-designed 3D-PRESS, allows precise pressure application up to 400 kPa at the single-cell level. Microfluidics is employed to encapsulate single mesenchymal stem cells within ionically cross-linked alginate microgels with cell adhesion RGD peptides. Rigorous testing affirms the leak-proof performance of the 3D-PRESS device up to 400 kPa, which is fully biocompatible. 3D-PRESS is implemented on mesenchymal stem cells for mechanotransduction studies, by specifically targeting intracellular calcium signaling and the nuclear translocation of a mechanically sensitive transcription factor. Applying 200 kPa pressure on individually encapsulated stem cells reveals heightened calcium signaling in 3D microgels compared to conventional 2D culture. Similarly, Yes-associated protein (YAP) translocation into the nucleus occurs at 200 kPa in 3D microgels with cell-binding RGD peptides unveiling the involvement of integrin-mediated mechanotransduction in singly encapsulated stem cells in 3D microgels. Combining live-cell imaging with precise mechanical control, the 3D-PRESS platform emerges as a versatile tool for exploring cellular responses to pressure stimuli, applicable to various cell types, providing novel insights into single-cell mechanobiology.
Collapse
Affiliation(s)
- Nergishan İyisan
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| | - Oliver Hausdörfer
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
| | - Chen Wang
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| | - Lukas Hiendlmeier
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
- Neuroelectronics, School of Computation, Information, and TechnologyDepartment of Electrical EngineeringDepartment of Electrical EngineeringTechnical University of Munich (TUM)85748GarchingGermany
| | - Philipp Harder
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| | - Bernhard Wolfrum
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
- Neuroelectronics, School of Computation, Information, and TechnologyDepartment of Electrical EngineeringDepartment of Electrical EngineeringTechnical University of Munich (TUM)85748GarchingGermany
| | - Berna Özkale
- Microrobotic Bioengineering Lab (MRBL)School of Computation, Information, and TechnologyDepartment of Electrical EngineeringTechnical University of Munich (TUM)Hans‐Piloty‐Straße 185748GarchingGermany
- Munich Institute of Robotics and Machine IntelligenceTechnical University of MunichGeorg‐Brauchle‐Ring 6080992MünchenGermany
- Munich Institute of Biomedical EngineeringTechnical University of MunichBoltzmannstraße 1185748GarchingGermany
| |
Collapse
|
23
|
Gao P, Kajiya M, Motoike S, Ikeya M, Yang J. Application of mesenchymal stem/stromal cells in periodontal regeneration: Opportunities and challenges. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:95-108. [PMID: 38314143 PMCID: PMC10837070 DOI: 10.1016/j.jdsr.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Guided tissue regeneration (GTR) has been widely used in the periodontal treatment of intrabony and furcation defects for nearly four decades. The treatment outcomes have shown effectiveness in reducing pocket depth, improving attachment gain and bone filling in periodontal tissue. Although applying GTR could reconstruct the periodontal tissue, the surgical indications are relatively narrow, and some complications and race ethic problems bring new challenges. Therefore, it is challenging to achieve a consensus concerning the clinical benefits of GTR. With the appearance of stem cell-based regenerative medicine, mesenchymal stem/stromal cells (MSCs) have been considered a promising cell resource for periodontal regeneration. In this review, we highlight preclinical and clinical periodontal regeneration using MSCs derived from distinct origins, including non-odontogenic and odontogenic tissues and induced pluripotent stem cells, and discuss the transplantation procedures, therapeutic mechanisms, and concerns to evaluate the effectiveness of MSCs.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Souta Motoike
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jingmei Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
24
|
Niebergall-Roth E, Dieter K, Frank MH, Kluth MA. Systemic treatment of recessive dystrophic epidermolysis bullosa with mesenchymal stromal cells: a scoping review of the literature and conclusions for future clinical research. J DERMATOL TREAT 2024; 35:2419931. [PMID: 39551482 DOI: 10.1080/09546634.2024.2419931] [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: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Background: The ability of mesenchymal stromal cells (MSCs) to facilitate regenerative responses in inflamed and injured tissues, coupled with preclinical data suggesting potential to restore defective collagen VII at the dermo-epidermal junction, has raised the hope that MSCs may provide an effective disease-modifying therapy for patients suffering from recessive dystrophic epidermolysis bullosa (RDEB). Methods: We present a descriptive analysis of the clinical research on systemic MSC administration to RDEB patients available in PubMed, including six early-phase studies and one case report, involving 59 patients who received 1-3 intravenous infusions of MSCs from various sources. Results: Based on 133 MSC infusions, a total of 44 mostly mild adverse events were reported as definitely, possibly or likely related to the study treatment, only two of which led to treatment discontinuation. Improvements were seen in skin manifestations, disease activity, pain, pruritus and quality of life, with considerable heterogeneity in reported outcome variables and measurement tools between studies, and large inter-patient variability within studies. Conclusions: Although the current evidence base is limited, reflecting the typical challenges of clinical research in rare diseases, the reported results suggest potential treatment benefits for patients and provide a rationale for continuing to pursue this therapeutic approach.
Collapse
Affiliation(s)
| | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | | |
Collapse
|
25
|
Gao Q, Cekuc MS, Ergul YS, Pius AK, Shinohara I, Murayama M, Susuki Y, Ma C, Morita M, Chow SKH, Goodman SB. 3D Culture of MSCs for Clinical Application. Bioengineering (Basel) 2024; 11:1199. [PMID: 39768017 PMCID: PMC11726872 DOI: 10.3390/bioengineering11121199] [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: 10/30/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) play an important role in regenerative medicine and drug discovery due to their multipotential differentiation capabilities and immunomodulatory effects. Compared with traditional 2D cultures of MSCs, 3D cultures of MSCs have emerged as an effective approach to enhance cell viability, proliferation, and functionality, and provide a more relevant physiological environment. Here, we review the therapeutic potential of 3D-cultured MSCs, highlighting their roles in tissue regeneration and repair and drug screening. We further summarize successful cases that apply 3D MSCs in modeling disease states, enabling the identification of novel therapeutic strategies. Despite these promising applications, we discuss challenges that remain in the clinical translation of 3D MSC technologies, including stability, cell heterogeneity, and regulatory issues. We conclude by addressing these obstacles and emphasizing the need for further research to fully exploit the potential of 3D MSCs in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94304, USA; (Q.G.)
| |
Collapse
|
26
|
Mera Azaín CA, Vargas Pasquel JL, Quijano Gómez SM, Rodríguez-Pardo VM. Mesenchymal Stem Cells and Reticulated Platelets: New Horizons in Multiple Myeloma. Hematol Rep 2024; 16:732-741. [PMID: 39584927 PMCID: PMC11627159 DOI: 10.3390/hematolrep16040070] [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: 09/18/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder characterized by the accumulation of abnormal plasma cells in the bone marrow. Mesenchymal stem cells (MSCs) and reticulated platelets (RPs) have been implicated in the pathogenesis of MM. This narrative review aims to explore the role of MSCs and RPs in the pathophysiology of MM, particularly their clinical use as possible variables of prognostic value in this hematologic neoplasia. The interaction between MSCs and MM cells within the bone marrow microenvironment supports MM cell survival, proliferation, and drug resistance. MSCs contribute to the development and maintenance of MM through the secretion of various factors, including cytokines, chemokines, and growth factors. Moreover, RPs, young and highly reactive platelets, have been implicated in promoting angiogenesis, tumor growth, and metastasis in MM. Several studies show that cells such as MSCs and platelets participate actively in the biology of the disease. Still, in clinical practice, they are not considered part of evaluating affected patients. In this review, we explore the possibility of including the evaluation of MSCs and PRs in the clinical practice for patients with MM as part of the strategies to improve the outcomes of this disease.
Collapse
Affiliation(s)
| | | | | | - Viviana Marcela Rodríguez-Pardo
- Grupo Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110111, Colombia; (C.A.M.A.); (J.L.V.P.); (S.M.Q.G.)
| |
Collapse
|
27
|
Barilani M, Peli V, Manzini P, Pistoni C, Rusconi F, Pinatel EM, Pischiutta F, Tace D, Iachini MC, Elia N, Tribuzio F, Banfi F, Sessa A, Cherubini A, Dolo V, Bollati V, Fiandra L, Longhi E, Zanier ER, Lazzari L. Extracellular Vesicles from Human Induced Pluripotent Stem Cells Exhibit a Unique MicroRNA and CircRNA Signature. Int J Biol Sci 2024; 20:6255-6278. [PMID: 39664576 PMCID: PMC11628337 DOI: 10.7150/ijbs.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Extracellular vesicles (EV) have emerged as promising cell-free therapeutics in regenerative medicine. However, translating primary cell line-derived EV to clinical applications requires large-scale manufacturing and several challenges, such as replicative senescence, donor heterogeneity, and genetic instability. To address these limitations, we used a reprogramming approach to generate human induced pluripotent stem cells (hiPSC) from the young source of cord blood mesenchymal stem/stromal cells (CBMSC). Capitalizing on their inexhaustible supply potential, hiPSC offer an attractive EV reservoir. Our approach encompassed an exhaustive characterization of hiPSC-EV, aligning with the rigorous MISEV2023 guidelines. Analyses demonstrated physical features compatible with small EV (sEV) and established their identity and purity. Moreover, the sEV-shuttled non-coding (nc) RNA landscape, focusing on the microRNA and circular RNA cargo, completed the molecular signature. The kinetics of the hiPSC-sEV release and cell internalization assays unveiled robust EV production and consistent uptake by human neurons. Furthermore, hiPSC-sEV demonstrated ex vivo cell tissue-protective properties. Finally, via bioinformatics, the potential involvement of the ncRNA cargo in the hiPSC-sEV biological effects was explored. This study significantly advances the understanding of pluripotent stem cell-derived EV. We propose cord blood MSC-derived hiPSC as a promising source for potentially therapeutic sEV.
Collapse
Affiliation(s)
- Mario Barilani
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valeria Peli
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Paolo Manzini
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Clelia Pistoni
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Medical Oncology and Hematology, University Hospital Zurich, Switzerland
| | - Francesco Rusconi
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Eva Maria Pinatel
- ITB-CNR, Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Francesca Pischiutta
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Dorian Tace
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Maria Chiara Iachini
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Noemi Elia
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Francesca Tribuzio
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Federica Banfi
- San Raffaele Scientific Institute, Division of Neuroscience, Neuroepigenetics Unit, Milano, Italy
| | - Alessandro Sessa
- San Raffaele Scientific Institute, Division of Neuroscience, Neuroepigenetics Unit, Milano, Italy
| | - Alessandro Cherubini
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
| | - Luisa Fiandra
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milano, Italy
| | - Elena Longhi
- Laboratory of Transplant Immunology SC Trapianti Lombardia - NITp. Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elisa R Zanier
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Lorenza Lazzari
- Unit of Cell and Gene Therapies, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| |
Collapse
|
28
|
Liu T, Ran C, Zhao D, Yang F, Guo Q, Yang J, Zhang X. Mesenchymal stem cells and their exosomes mitigate osteoarthritis by restoring the balance between proinflammatory Teffs and Tregs. FRONTIERS IN AGING 2024; 5:1509014. [PMID: 39629263 PMCID: PMC11611854 DOI: 10.3389/fragi.2024.1509014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease caused by chronic inflammation that damages articular cartilage. In addition to the wear and tear of joints, aberrant remodelling driven by a significant presence of inflammatory mediators within the joint is one of the key mechanisms in the pathogenesis of OA. Among these factors, hyperactivation of Teffs subsets plays a crucial role in promoting this pathological process. The immune imbalance between proinflammatory CD4+ effector T cells (proinflammatory Teffs) and Tregs could be a crucial factor in the pathogenesis of OA. Therefore, correcting the imbalance of Tregs/proinflammatory Teffs may slow or inhibit the occurrence and development of OA, which could be a potential target for the treatment of OA. Mesenchymal stem cells (MSCs) possess anti-inflammatory and immunomodulatory properties, regulating both adaptive and innate immunity through mechanisms involving soluble factors such as IDO, PGE2, and TGF-β, as well as cell-to-cell contact and exosomes. Correcting the imbalance between Tregs and proinflammatory Teffs may be one of the mechanisms of MSCs in the treatment of OA. Therefore, this review aims to summarize the relationship between OA and the immune imbalance between Tregs and proinflammatory Teffs, the immunoregulatory role of Tregs in OA, and the role of MSCs and their exosomes in correcting the imbalance between Tregs and proinflammatory Teffs.
Collapse
Affiliation(s)
- Tianhao Liu
- Zhongshan Clinical College, Dalian University, Dalian, Liaoning, China
| | - Chunxiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dewei Zhao
- Zhongshan Clinical College, Dalian University, Dalian, Liaoning, China
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Fan Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Qiang Guo
- Zhongshan Clinical College, Dalian University, Dalian, Liaoning, China
| | - Jiahui Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xiuzhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
29
|
Khaveh N, Buschow R, Metzger J. Deciphering transcriptome patterns in porcine mesenchymal stem cells promoting phenotypic maintenance and differentiation by key driver genes. Front Cell Dev Biol 2024; 12:1478757. [PMID: 39568509 PMCID: PMC11576426 DOI: 10.3389/fcell.2024.1478757] [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: 08/10/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Mesenchymal stem cells (MSC) are fibroblast-like non-hematopoietic cells with self-renewal and differentiation capacity, and thereby great potential in regeneration and wound healing. MSC populations are heterogeneous not only inherently, but also among different model species. In particular, porcine MSC serve as a frequently used resource for translational research, due to pigs' distinctive closeness to human anatomy and physiology. However, information on gene expression profiles from porcine MSC and its dynamics during differentiation is sparse, especially with regard to cell surface and inner cell markers. In this study, we investigated the transcriptome of bone marrow-derived MSC and its differentiated cell types in a minipig breed for experimental research, known as Mini-LEWE, using bulk mRNA sequencing. Our data highlighted Rap1 signaling and downstream pathways PI3K-Akt and MAPK signaling as potential players for the maintenance of stemness of BM-MSC. In addition, we were able to link the process of differentiation to changes in the regulation of actin cytoskeleton. A total of 18 "BM-MSC differentiation driver markers" were identified, potentially promoting the process of differentiation into adipocytes, chondrocytes as well as osteocytes. Our results offer a new perspective on the molecular phenotype of porcine BM-MSC and the transcriptional responses in new differentiated progeny.
Collapse
Affiliation(s)
- Nadia Khaveh
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - René Buschow
- Microscopy and Cryo-Electron Microscopy Facility, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Julia Metzger
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Group Veterinary Functional Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
30
|
Campbell JM, Habibalahi A, Agha A, Handley S, Knab A, Xu X, Bhargava A, Lei Z, Mackevicius M, Tian Y, Mahbub SB, Anwer AG, Gronthos S, Paton S, Grey ST, Wu L, Gilchrist RB, Goldys EM. Single cell, Label free Characterisation of Human Mesenchymal Stromal cell Stemness and Future Growth Potential by Autofluorescence Multispectral Imaging. Stem Cell Rev Rep 2024; 20:2283-2292. [PMID: 39190057 PMCID: PMC11554749 DOI: 10.1007/s12015-024-10778-4] [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] [Accepted: 08/15/2024] [Indexed: 08/28/2024]
Abstract
AIM To use autofluorescence multispectral imaging (AFMI) to develop a non-invasive assay for the in-depth characterisation of human bone marrow derived mesenchymal stromal cells (hBM-MSCs). METHODS hBM-MSCs were imaged by AFMI on gridded dishes, stained for endpoints of interest (STRO-1 positivity, alkaline phosphatase, beta galactosidase, DNA content) then relocated and results correlated. Intensity, texture and morphological features were used to characterise the colour distribution of regions of interest, and canonical discriminant analysis was used to separate groups. Additionally, hBM-MSC lines were cultured to arrest, with AFMI images taken after each passage to investigate whether an assay could be developed for growth potential. RESULTS STRO-1 positivity could be predicted with a receiver operator characteristic area under the curve (AUC) of 0.67. For spontaneous differentiation this was 0.66, for entry to the cell-cycle it was 0.77 and for senescence it was 0.77. Growth potential (population doublings remaining) was estimated with an RMSPE = 2.296. The Mean Absolute Error of the final prediction model indicated that growth potential could be predicted with an error of ± 1.86 doublings remaining. CONCLUSIONS This non-invasive methodology enabled the in-depth characterisation of hBM-MSCs from a single assay. This approach is advantageous for clinical applications as well as research and stands out for the characterisation of both present status as well as future behaviour. The use of data from five MSC lines with heterogenous AFMI profiles supports potential generalisability.
Collapse
Affiliation(s)
- Jared M Campbell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Abbas Habibalahi
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Adnan Agha
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shannon Handley
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Aline Knab
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Xiaohu Xu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Akanksha Bhargava
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zhilin Lei
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Max Mackevicius
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Yuan Tian
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Saabah B Mahbub
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Ayad G Anwer
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia
| | - Sharon Paton
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia
| | - Shane T Grey
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lindsay Wu
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | | | - Ewa M Goldys
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
31
|
Sun S, Wang L, Tang Q, Yi J, Yu X, Cao Y, Jiang L, Liu J. Myocardial infarction in rats was alleviated by MSCs derived from the maternal segment of the human umbilical cord. Front Cell Dev Biol 2024; 12:1469541. [PMID: 39479514 PMCID: PMC11521943 DOI: 10.3389/fcell.2024.1469541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) are safe and effective in treating myocardial infarction (MI) and have broad application prospects. However, the heterogeneity of MSCs may affect their therapeutic effect on the disease. We recently found that MSCs derived from different segments of the same umbilical cord (UC) showed significant difference in the expression of genes that are related to heart development and injury repair. We therefore hypothesized that those MSCs with high expression of above genes are more effective to treat MI and tested it in this study. Methods MSCs were isolated from 3 cm-long segments of the maternal, middle and fetal segments of the UC (maternal-MSCs, middle-MSCs and fetal-MSCs, respectively). RNA-seq was used to analyze and compare the transcriptomes. We verified the effects of MSCs on oxygen-glucose deprivation (OGD)-induced cardiomyocyte apoptosis in vitro. In vivo, a rat MI model was established by ligating the left anterior descending coronary artery, and MSCs were injected into the myocardium surrounding the MI site. The therapeutic effects of MSCs derived from different segments of the UC were evaluated by examining cardiac function, histopathology, cardiomyocyte apoptosis, and angiogenesis. Results Compared to fetal-MSCs and middle-MSCs, maternal-MSCs exhibited significantly higher expression of genes that are associated with heart development, such as GATA-binding protein 4 (GATA4), and myocardin (MYOCD). Coculture with maternal-MSCs reduced OGD-induced cardiomyocyte apoptosis. In rats with MI, maternal-MSCs significantly restored cardiac contractile function and reduced the infarct size. Mechanistic experiments revealed that maternal-MSCs exerted cardioprotective effects by decreasing cardiomyocyte apoptosis, and promoting angiogenesis. Conclusion Our data demonstrated that maternal segment-derived MSCs were a superior cell source for regenerative repair after MI. Segmental localization of the entire UC when isolating hUCMSCs was necessary to improve the effectiveness of clinical applications.
Collapse
Affiliation(s)
- Shuifen Sun
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Linping Wang
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Qisheng Tang
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Jialian Yi
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Xin Yu
- Medicine School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yu Cao
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Lihong Jiang
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jie Liu
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
32
|
Bzinkowska A, Sarnowska A. Assessment of the Dose-Dependent Effect of Human Platelet Lysate on Wharton's Jelly-Derived Mesenchymal Stem/Stromal Cells Culture for Manufacturing Protocols. Stem Cells Cloning 2024; 17:21-32. [PMID: 39386994 PMCID: PMC11463174 DOI: 10.2147/sccaa.s471118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Mesenchymal stem/stromal cells (MSCs)-based products have unique characteristics compared to other drugs because of their inherently variable effects depending on culture conditions and microenvironment. In some cases, cells can be produced individually, one batch at a time, for personalized therapy. Therefore, it is very important to optimize both culture conditions and medium composition under Good Manufacturing Practice (GMP) standards. MSCs properties have been exploited as potential cell therapies in regenerative medicine. The main mechanism of their protective and regenerative effect is based on their secretory activity. Simultaneously, their secretome is highly variable and sensitive to any change in environmental conditions. Depending on the type of damage and the target application, it is desirable to enhance the secretion of therapeutic factors. Changes in the modulation of environmental conditions can affect survival, migration ability, and both proliferative and clonogenic potentials. Materials and Methods This study cultured Wharton's jelly-derived MSCs (WJ-MSCs) in media with varying concentrations of human platelet lysate (hPL). Two groups were created: one with low hPL concentration and another with a high hPL concentration. The effects of these different hPL concentrations were analyzed by assessing mesenchymal phenotype retention, secretory activity, clonogenic potential, proliferation, and migration capabilities. Additionally, the secretion levels of key therapeutic factors, such as Hepatocyte Growth Factor (HGF), Brain-Derived Neurotrophic Factor (BDNF), and Chemokine Ligand 2 (CCL-2), were measured. Results WJ-MSCs maintained their mesenchymal phenotype regardless of hPL concentration. However, a higher concentration of hPL promoted cell clonogenic potential, proliferation, migration, and increased secretion of therapeutic factors. Conclusion Adjusting the hPL concentration in the culture medium modulates the response of WJ MSCs and enhances their therapeutic potential. Higher hPL concentration promotes increased secretory activity and improves the regenerative capacity of WJ-MSCs, suggesting a promising strategy to optimize MSC-based therapies.
Collapse
Affiliation(s)
- Aleksandra Bzinkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
33
|
Kang M, Yang Y, Zhang H, Zhang Y, Wu Y, Denslin V, Othman RB, Yang Z, Han J. Comparative Analysis of Serum and Serum-Free Medium Cultured Mesenchymal Stromal Cells for Cartilage Repair. Int J Mol Sci 2024; 25:10627. [PMID: 39408956 PMCID: PMC11476526 DOI: 10.3390/ijms251910627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for cartilage repair therapy due to their self-renewal, chondrogenic, and immunomodulatory capacities. It is widely recognized that a shift from fetal bovine serum (FBS)-containing medium toward a fully chemically defined serum-free (SF) medium would be necessary for clinical applications of MSCs to eliminate issues such as xeno-contamination and batch-to-batch variation. However, there is a notable gap in the literature regarding the evaluation of the chondrogenic ability of SF-expanded MSCs (SF-MSCs). In this study, we compared the in vivo regeneration effect of FBS-MSCs and SF-MSCs in a rat osteochondral defect model and found poor cartilage repair outcomes for SF-MSCs. Consequently, a comparative analysis of FBS-MSCs and SF-MSCs expanded using two SF media, MesenCult™-ACF (ACF), and Custom StemPro™ MSC SFM XenoFree (XF) was conducted in vitro. Our results show that SF-expanded MSCs constitute variations in morphology, surface markers, senescence status, differentiation capacity, and senescence/apoptosis status. Highly proliferative MSCs supported by SF medium do not always correlate to their chondrogenic and cartilage repair ability. Prior determination of the SF medium's ability to support the chondrogenic ability of expanded MSCs is therefore crucial when choosing an SF medium to manufacture MSCs for clinical application in cartilage repair.
Collapse
Affiliation(s)
- Meiqi Kang
- Critical Analytics for Manufacturing Personalised-Medicine (CAMP) Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore; (M.K.); (Y.Y.); (R.B.O.)
| | - Yanmeng Yang
- Critical Analytics for Manufacturing Personalised-Medicine (CAMP) Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore; (M.K.); (Y.Y.); (R.B.O.)
| | - Haifeng Zhang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore; (H.Z.); (Y.Z.); (Y.W.); (V.D.)
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore 117510, Singapore
| | - Yuan Zhang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore; (H.Z.); (Y.Z.); (Y.W.); (V.D.)
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore 117510, Singapore
| | - Yingnan Wu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore; (H.Z.); (Y.Z.); (Y.W.); (V.D.)
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore 117510, Singapore
| | - Vinitha Denslin
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore; (H.Z.); (Y.Z.); (Y.W.); (V.D.)
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore 117510, Singapore
| | - Rashidah Binte Othman
- Critical Analytics for Manufacturing Personalised-Medicine (CAMP) Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore; (M.K.); (Y.Y.); (R.B.O.)
| | - Zheng Yang
- Critical Analytics for Manufacturing Personalised-Medicine (CAMP) Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore; (M.K.); (Y.Y.); (R.B.O.)
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore; (H.Z.); (Y.Z.); (Y.W.); (V.D.)
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore 117510, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing Personalised-Medicine (CAMP) Interdisciplinary Research Group (IRG), Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore 138602, Singapore; (M.K.); (Y.Y.); (R.B.O.)
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| |
Collapse
|
34
|
Behm C, Miłek O, Schwarz K, Kovar A, Derdak S, Rausch-Fan X, Moritz A, Andrukhov O. Heterogeneity in Dental Tissue-Derived MSCs Revealed by Single-Cell RNA-seq. J Dent Res 2024; 103:1141-1152. [PMID: 39327720 PMCID: PMC11500480 DOI: 10.1177/00220345241271997] [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] [Indexed: 09/28/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent, progenitor cells that reside in tissues across the human body, including the periodontal ligament (PDL) and gingiva. They are a promising therapeutic tool for various degenerative and inflammatory diseases. However, different heterogeneity levels caused by tissue-to-tissue and donor-to-donor variability, and even intercellular differences within a given MSCs population, restrict their therapeutic potential. There are considerable efforts to decipher these heterogeneity levels using different "omics" approaches, including single-cell transcriptomics. Previous studies applied this approach to compare MSCs isolated from various tissues of different individuals, but distinguishing between donor-to-donor and tissue-to-tissue variability is still challenging. In this study, MSCs were isolated from the PDL and gingiva of 5 periodontally healthy individuals and cultured in vitro. A total of 3,844 transcriptomes were generated using single-cell mRNA sequencing. Clustering across the 2 different tissues per donor identified PDL- and gingiva-specific and tissue-spanning MSCs subpopulations with unique upregulated gene sets. Gene/pathway enrichment and protein-protein interaction (PPI) network analysis revealed differences restricted to several cellular processes between tissue-specific subpopulations, indicating a limited tissue-of-origin variability in MSCs. Gene expression, pathway enrichment, and PPI network analysis across all donors' PDL- or gingiva-specific subpopulations showed significant but limited donor-to-donor differences. In conclusion, this study demonstrates tissue- and donor-specific variabilities in the transcriptome level of PDL- and gingiva-derived MSCs, which seem restricted to specific cellular processes. Identifying tissue-specific and tissue-spanning subpopulations highlights the intercellular differences in dental tissue-derived MSCs. It could be reasonable to control MSCs at a single-cell level to ensure their properties before transplantation.
Collapse
Affiliation(s)
- C. Behm
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Austria
| | - O. Miłek
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Austria
| | - K. Schwarz
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Austria
| | - A. Kovar
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Austria
| | - S. Derdak
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - X. Rausch-Fan
- Clinical Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Austria
- Center for Clinical Research, University Clinic of Dentistry, Medical University of Vienna, Austria
| | - A. Moritz
- Clinical Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Austria
| | - O. Andrukhov
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Austria
| |
Collapse
|
35
|
Amiri F, Mistriotis P. Leveraging Cell Migration Dynamics to Discriminate Between Senescent and Presenescent Human Mesenchymal Stem Cells. Cell Mol Bioeng 2024; 17:385-399. [PMID: 39513008 PMCID: PMC11538215 DOI: 10.1007/s12195-024-00807-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/11/2024] [Indexed: 11/15/2024] Open
Abstract
Purpose The suboptimal clinical performance of human mesenchymal stem cells (hMSCs) has raised concerns about their therapeutic potential. One major contributing factor to this issue is the heterogeneous nature of hMSCs. Senescent cell accumulation during stem cell expansion is a key driver of MSC heterogeneity. Current methodologies to eradicate senescent hMSCs have either shown limited success or lack clinical relevance. This study leverages the inherent capacity of hMSCs to migrate toward damaged tissues as a means to discern senescent from presenescent stem cells. Given the established deficiency of senescent cells to migrate through physiologically relevant environments, we hypothesized that a microfluidic device, designed to emulate key facets of in vivo cell motility, could serve as a platform for identifying presenescent cells. Methods We employed a Y-shaped microchannel assay, which allows fine-tuning of fluid flow rates and the degree of confinement. Results Highly migratory hMSCs detected by the device not only demonstrate increased speed, smaller size, and higher proliferative capacity but also manifest reduced DNA damage and senescence compared to non-migratory cells. Additionally, this assay detects presenescent cells in experiments with mixed early and late passage cells. The introduction of fluid flow through the device can further increase the fraction of highly motile stem cells, improving the assay's effectiveness to remove senescent hMSCs. Conclusions Collectively, this assay facilitates the detection and isolation of a highly potent stem cell subpopulation. Given the positive correlation between the migratory potential of administered MSCs and the long-term clinical outcome, delivering homogeneous, highly motile presenescent hMSCs may benefit patient outcomes. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00807-0.
Collapse
Affiliation(s)
- Farshad Amiri
- Department of Chemical Engineering, Auburn University, Auburn, AL USA
| | | |
Collapse
|
36
|
Wang P, Zhang Y, Li Z, Zhou S, Tang Q, Wang Z, Xiao R, Feng M, Wu L, Liang D. Mesenchymal Stem Cells Derived from Human Urine-Derived iPSCs Exhibit Low Immunogenicity and Reduced Immunomodulatory Profile. Int J Mol Sci 2024; 25:10394. [PMID: 39408724 PMCID: PMC11476417 DOI: 10.3390/ijms251910394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Human-induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) represent a promising and renewable cell source for therapeutic applications. A systematic evaluation of the immunological properties and engraftment potential of iMSCs generated from urine-derived iPSCs is lacking, which has impeded their broader application. In this study, we differentiated urine-derived iPSCs into iMSCs and assessed their fundamental MSC characteristics, immunogenicity, immunomodulatory capacity and in vivo engraftment. Compared to umbilical cord-derived MSCs (UCMSCs), iMSCs demonstrated an enhanced proliferative capacity, a higher level of regenerative gene expression, and lower immunogenicity, strengthening resistance to apoptosis induced by allogeneic peripheral blood mononuclear cells (PBMCs) and the NK-92 cell line. In addition, iMSCs exhibited a diminished ability to inhibit T cell proliferation and activation compared with UCMSCs. Transcriptomic analyses further revealed the decreased expression of immune regulatory factors in iMSCs. After transfusion into mouse models, iMSCs engrafted in the lungs, liver, and spleen and exhibited the ability to migrate to tumor tissues. Our results indicated that iMSCs generated from urine-derived iPSCs have a significant replicative capacity, low immunogenicity and unique immunomodulatory properties, and hence offer obvious advantages in immune privilege and allogenic therapeutic application prospects.
Collapse
Affiliation(s)
- Peiyun Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Ying Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Zhixing Li
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Shenglan Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Qiyu Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Zujia Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Rou Xiao
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Mai Feng
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410078, China;
| | - Lingqian Wu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Desheng Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| |
Collapse
|
37
|
Castro Ramos A, Widjaja Lomanto MY, Vuong CK, Ohneda O, Fukushige M. Antibacterial effects of human mesenchymal stem cells and their derivatives: a systematic review. Front Microbiol 2024; 15:1430650. [PMID: 39386372 PMCID: PMC11461301 DOI: 10.3389/fmicb.2024.1430650] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/12/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction The growing problem of antimicrobial resistance (AMR) poses a significant challenge to public health; This is partly due to the lack of advancements in the development of novel antibiotics and the pressing need for alternative treatment options. Mesenchymal stem cells (MSC) possess secretory components that enhance the immune response and peptides that disrupt the bacteria constitution. The isolation of various human tissues has facilitated the investigation of the diverse potentials of MSC and their components. Further research is needed to fully understand the spectrum and efficacy of these elements and their differences. The primary aim of this study was to perform a thorough review of the current literature related to the antimicrobial properties of MSC and their associated components. The objective was to establish an insight into the results and effects of utilizing MSC in relation to bacterial colonization, and to present an overview of previously documented findings. Methods This systematic literature review was conducted using the PubMed, Embase, and Web of Science databases. Data on the effect of MSC or their derivatives were measured by calculating the percentage of bacterial counts reduction after treatment with MSC in comparison to the control. Results A total of 3,911 articles were screened, and 31 eligible publications were selected for inclusion in the analysis. In the current systematic review, the majority of the experimental designs showed positive outcomes in terms of bacterial load reduction when MSC or their derivatives were used, with bone marrow being the most effective tissue. The rest of the findings exhibited heterogeneity in the spectrum of outcomes that could be attributed to the effects of using various tissues derived MSC in both in vivo and in vitro studies. Conclusion The findings of our study indicate the potential antibacterial characteristics of MSC. The direct antimicrobial activity of these cells was demonstrated by our results, which quantitatively showed a decrease in bacterial growth after treatment with MSC. However, additional research is required to clarify the factors that determine the efficacy of their antimicrobial activity and their various components.
Collapse
Affiliation(s)
- Adeline Castro Ramos
- Graduate School of Comprehensive Human Science, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Markus Yovian Widjaja Lomanto
- School of Integrative and Global Majors, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Cat-Khanh Vuong
- Laboratory of Regenerative Medicine and Stem Cell Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Osamu Ohneda
- Laboratory of Regenerative Medicine and Stem Cell Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Mizuho Fukushige
- Laboratory of Regenerative Medicine and Stem Cell Biology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
38
|
Fadaghi S, Mahmoodi M, Derakhshani A, Sedghy F, Ranjkesh M, Behzadi A. Enhancement the antioxidative and immunomodulatory functions of mesenchymal stem cells by tetrandrine. Heliyon 2024; 10:e35667. [PMID: 39220890 PMCID: PMC11365297 DOI: 10.1016/j.heliyon.2024.e35667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
In this study, mesenchymal stem cells (MSCs) were primed with Tetrandrine (TET) having anti-inflammatory and immunomodulatory effects to examine the effects of this molecule on the antioxidative potential of MSCs as well as their modulatory effects on activated peripheral blood mononuclear cells (PBMCs). The viability of primed MSCs was detected using MTT assay and Trypan blue staining. Moreover, flow cytometry technique was applied to evaluate cell cycle distribution and immunophenotype of MSCs. The production of superoxide dismutase 3 (SOD3), malondialdehyde (MDA), kynurenine, TGF-β, and IFN-γ were also measured by spectrophotometry to assess the alteration of antioxidative and immunomodulatory potential of MSCs. Then, TET-primed MSCs were cocultured with PBMCs. The MTT assay was used to measure the proliferation of PBMCs. Cell cycle progression of PBMCs and frequency of regulatory T cells were evaluated using Flow cytometry. ELISA assay was also applied to determine the concentrations of TGF-β and IFN-γ after coculturing. According to our data, TET enhanced the secretion of SOD3 and kynurenine from MSCs, while the production of IFN-γ was reduced. No changes were observed in the viability, proliferation, and immunophenotype of MSCs after priming with TET. Moreover, the proliferation and frequency of PBMCs in the S and G2/M phases of cell cycle reduced after co-culturing with TET-primed MSCs. The concentration of TGF-β was increased in the supernatant of PBMCs, but the level of IFN-γ was reduced. Our data suggested this priming method as a novel strategy for increasing the antioxidative and immunomodulatory activity of MSCs.
Collapse
Affiliation(s)
- Shohreh Fadaghi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Merat Mahmoodi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Derakhshani
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Farnaz Sedghy
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdi Ranjkesh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmadreza Behzadi
- School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
39
|
Khastar S, Sattari M. Examining the level of inflammatory cytokines TNF-α and IL-8 produced by osteoblasts differentiated from dental pulp stem cells. AMERICAN JOURNAL OF STEM CELLS 2024; 13:225-232. [PMID: 39308765 PMCID: PMC11411251 DOI: 10.62347/cbmw4366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND The use of dental pulp stem cells (DPSCs) in clinical applications instead of bone marrow stem cells is a very promising method capable of significantly changing the future of medical treatment. If further studies prove that DPSCs and the cells differentiated from them do not stimulate the immune system, these cells can be used more reliably in treatment of autoimmune diseases. METHODS In this research, we examined the isolated DPSCs and differentiated osteoblasts from them in medium without inflammatory stimulants in terms of TLR3 and TLR4 gene expression and inflammatory cytokines, including TNF-α and IL-8 using qRT-PCR, and measured the concentration of inflammatory cytokines IL-8 and TNF-α produced by these two types of cells through ELISA. RESULTS The obtained results showed that the expression level of inflammatory cytokines IL-8 and TNF-α in differentiated osteoblasts is significantly different as compared with DPSCs. However, no significant difference was observed in TLR-4 expression between two groups. An increase in TNF-α expression level was found to directly correlate with an increase in the expression of IL-8. The concentration of cytokine TNF-α in osteoblasts was significantly higher than that of IL-8 in DPSCs. CONCLUSION In comparison to DPSCs, osteoblast cells first lead to inflammatory responses. These responses reduce overtime. However, DPSCs retain their immunomodulatory properties and do not show inflammatory responses.
Collapse
Affiliation(s)
- Sahar Khastar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Mandana Sattari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences Tehran, Iran
| |
Collapse
|
40
|
Hisamatsu D, Ikeba A, Yamato T, Mabuchi Y, Watanabe M, Akazawa C. Optimization of transplantation methods using isolated mesenchymal stem/stromal cells: clinical trials of inflammatory bowel diseases as an example. Inflamm Regen 2024; 44:37. [PMID: 39152520 PMCID: PMC11328379 DOI: 10.1186/s41232-024-00350-5] [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: 04/24/2024] [Accepted: 07/26/2024] [Indexed: 08/19/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are distributed in various tissues and are used in clinical applications as a source of transplanted cells because of their easy harvestability. Although MSCs express numerous cell-surface antigens, single-cell analyses have revealed a highly heterogeneous cell population depending on the original tissue and donor conditions, including age and interindividual differences. This heterogeneity leads to differences in their functions, such as multipotency and immunomodulatory effects, making it challenging to effectively treat targeted diseases. The therapeutic efficacy of MSCs is controversial and depends on the implantation site. Thus, there is no established recipe for the transplantation of MSCs (including the type of disease, type of origin, method of cell culture, form of transplanted cells, and site of delivery). Our recent preclinical study identified appropriate MSCs and their suitable transplantation routes in a mouse model of inflammatory bowel disease (IBD). Three-dimensional (3D) cultures of MSCs have been demonstrated to enhance their properties and sustain engraftment at the lesion site. In this note, we explore the methods of MSC transplantation for treating IBDs, especially Crohn's disease, from clinical trials published over the past decade. Given the functional changes in MSCs in 3D culture, we also investigate the clinical trials using 3D constructs of MSCs and explore suitable diseases that might benefit from this approach. Furthermore, we discuss the advantages of the prospective isolation of MSCs in terms of interindividual variability. This note highlights the need to define the method of MSC transplantation, including interindividual variability, the culture period, and the transplantation route.
Collapse
Affiliation(s)
- Daisuke Hisamatsu
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Akimi Ikeba
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Taku Yamato
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Yo Mabuchi
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Tokyo, Japan
| | - Mamoru Watanabe
- Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Chihiro Akazawa
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
41
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
42
|
Chopra H, Cao C, Alice H, Kak S, Maska B, Tagett R, Sugai J, Garmire L, Kaigler D. Landscape of Differentiation Potentials as a "Hallmark" in Oral-derived MSCs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606413. [PMID: 39211139 PMCID: PMC11360929 DOI: 10.1101/2024.08.02.606413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Mesenchymal stem cells (MSCs) offer clinical promise for use in cell therapy approaches for regenerative medicine. A therapeutic challenge is that MSCs from different tissues are phenotypically and functionally distinct. Therefore, this study aims to molecularly characterize oral-derived MSCs by defining one of the three hallmarks of MSCs, differentiation potential, to discern their true molecular identities. Methods Three different populations of oral tissue MSCs (from alveolar bone-aBMSCs; from dental pulp-DPSCs; and from gingiva-GMSCs) from three different patients were isolated and cultured. These MSCs were characterized for their stemness by flow cytometry and multi-differentiation potential, and their RNA was also isolated and analyzed quantitatively with RNA sequencing. Total mRNA-seq was performed and differentially expressed genes (DEGs) were identified in pairwise (DPSCs vs. aBMSCs, GMSCs vs. aBMSCs, and GMSCs vs. DPSCs) and tissue-specific comparisons (aBMSCs vs. Others, DPSCs vs. Others, GMSCs vs. Others) (FDR, p<0.05 ). Further, these DEGs, either common between MSC populations or unique to a specific MSC population, were evaluated for pathways and biological processes. Results aBMSCs, DPSCs, and GMSCs were successfully isolated and characterized. The tissue-specific comparison revealed that DEGs were most numerous in DPSCs (693 genes) as compared to aBMSCs (103 genes) or DPSCs (232 genes). Statistically significant DEGs through pairwise comparisons present higher numbers in GMSCs vs. DPSCs (627) as compared to either DPSCs vs aBMSCs (286) or GMSCs vs. aBMSCs (82). Further analysis found that RUNX2, IBSP, SOX6, ACAN, and VCAM1 were significantly upregulated in aBMSCs. In DPSCs, BMP4 and IL6 were significantly downregulated, whereas AXL and NES were significantly upregulated. In GMSCs, AGPT1, SEMA4D, and PGDFA were significantly downregulated. Additionally, MAPK, PI3-AKT, and RAS signaling pathways were significantly regulated in GMSCs. Interestingly, aBMSCs and DPSCs revealed positive regulation of osteoblast differentiation, whereas GMSCs revealed negative regulation of osteoblast differentiation. DPSCs also revealed negative regulation of angiogenesis. Conclusions Oral-derived MSCs have an inherent "landscape" of differentiation defined by their tissue of origin; yet this differentiation potential can be modulated by their microenvironment.
Collapse
|
43
|
Lee SB, Abdal Dayem A, Kmiecik S, Lim KM, Seo DS, Kim HT, Kumar Biswas P, Do M, Kim DH, Cho SG. Efficient improvement of the proliferation, differentiation, and anti-arthritic capacity of mesenchymal stem cells by simply culturing on the immobilized FGF2 derived peptide, 44-ERGVVSIKGV-53. J Adv Res 2024; 62:119-141. [PMID: 37777063 PMCID: PMC11331723 DOI: 10.1016/j.jare.2023.09.041] [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: 09/06/2022] [Revised: 08/23/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
INTRODUCTION The stem cell microenvironment has been evidenced to robustly affect its biological functions and clinical grade. Natural or synthetic growth factors, especially, are essential for modulating stem cell proliferation, metabolism, and differentiation via the interaction with specific extracellular receptors. Fibroblast growth factor-2 (FGF-2) possesses pleiotropic functions in various tissues and organs. It interacts with the FGF receptor (FGFR) and activates FGFR signaling pathways, which involve numerous biological functions, such as angiogenesis, wound healing, cell proliferation, and differentiation. OBJECTIVES Here, we aim to explore the molecular functions, mode of action, and therapeutic activity of yet undetermined function, FGF-2-derived peptide, FP2 (44-ERGVVSIKGV-53) in promoting the proliferation, differentiation, and therapeutic application of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) in comparison to other test peptides, canofin1 (FP1), hexafin2 (FP3), and canofin3 (FP4) with known functions. METHODS The immobilization of test peptides that are fused with mussel adhesive proteins (MAP) on the culture plate was carried out via EDC/NHS chemistry. Cell Proliferation assay, colony-forming unit, western blotting analysis, gene expression analysis, RNA-Seq. analysis, osteogenic, and chondrogenic differentiation capacity were applied to test the activity of the test peptides. We additionally utilized three-dimensional (3D) structural analysis and artificial intelligence (AI)-based AlphaFold2 and CABS-dock programs for receptor interaction prediction of the peptide receptor. We also verified the in vivo therapeutic capacity of FP2-cultured hWJ-MSCs using an osteoarthritis mice model. RESULTS Culture of hWJ-MSC onto an FP2-immobilized culture plate showed a significant increase in cell proliferation (n = 3; *p < 0.05, **p < 0.01) and the colony-forming unit (n = 3; *p < 0.05, **p < 0.01) compared with the test peptides. FP2 showed a significantly upregulated phosphorylation of FRS2α and FGFR1 and activated the AKT and ERK signaling pathways (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). Interestingly, we detected efficient FP2 receptor binding that was predicted using AI-based tools. Treatment with an AKT inhibitor significantly abrogated the FP2-mediated enhancement of cell differentiation (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). Intra-articular injection of FP2-cultured MSCs significantly mitigated arthritis symptoms in an osteoarthritis mouse model, as shown through the functional tests (n = 10; *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001), modulation of the expression level of the pro-inflammatory and anti-inflammatory genes, and improved osteochondral regeneration as demonstrated by tissue sections. CONCLUSION Our study identified the FGF-2-derived peptide FP2 as a promising candidate peptide to improve the therapeutic potential of hWJ-MSCs, especially in bone and cartilage regeneration.
Collapse
Affiliation(s)
- Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sebastian Kmiecik
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-089 Warsaw, Poland
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong Sik Seo
- Stem Cell Research Center of AMOLIFESCIENCE Co., Ltd, 91, Gimpo-daero 1950 Beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Hyeong-Taek Kim
- Stem Cell Research Center of AMOLIFESCIENCE Co., Ltd, 91, Gimpo-daero 1950 Beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Polash Kumar Biswas
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
44
|
Kaur G, Bae E, Zhang Y, Ciacciofera N, Jung KM, Barreda H, Paleti C, Oh JY, Lee RH. Biopotency and surrogate assays to validate the immunomodulatory potency of extracellular vesicles derived from mesenchymal stem/stromal cells for the treatment of experimental autoimmune uveitis. J Extracell Vesicles 2024; 13:e12497. [PMID: 39140452 PMCID: PMC11322862 DOI: 10.1002/jev2.12497] [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: 08/23/2023] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) have been recognized as promising cytotherapeutics due to their demonstrated immunomodulatory effects in various preclinical models. The immunomodulatory capabilities of EVs stem from the proteins and genetic materials they carry from parent cells, but the cargo contents of EVs are significantly influenced by MSC tissues and donors, cellular age and culture conditions, resulting in functional variations. However, there are no surrogate assays available to validate the immunomodulatory potency of MSC-EVs before in vivo administration. In previous work, we discovered that microcarrier culture conditions enhance the immunomodulatory function of MSC-EVs, as well as the levels of immunosuppressive molecules such as TGF-β1 and let-7b in MSC-EVs. Building on these findings, we investigated whether TGF-β1 levels in MSC-EVs could serve as a surrogate biomarker for predicting their potency in vivo. Our studies revealed a strong correlation between TGF-β1 and let-7b levels in MSC-EVs, as well as their capacity to suppress IFN-γ secretion in stimulated splenocytes, establishing biopotency and surrogate assays for MSC-EVs. Subsequently, we validated MSC-EVs generated from monolayer cultures (ML-EVs) or microcarrier cultures (MC-EVs) using murine models of experimental autoimmune uveoretinitis (EAU) and additional in vitro assays reflecting the Mode of Action of MSC-EVs in vivo. Our findings demonstrated that MC-EVs carrying high levels of TGF-β1 exhibited greater efficacy than ML-EVs in halting disease progression in mice with EAU as well as inducing apoptosis and inhibiting the chemotaxis of retina-reactive T cells. Additionally, MSC-EVs suppressed the MAPK/ERK pathway in activated T cells, with treatment using TGF-β1 or let-7b showing similar effects on the MAPK/ERK pathway. Collectively, our data suggest that MSC-EVs directly inhibit the infiltration of retina-reactive T cells toward the eyes, thereby halting the disease progression in EAU mice, and their immunomodulatory potency in vivo can be predicted by their TGF-β1 levels.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Eun‐Hye Bae
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Yu Zhang
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Nicole Ciacciofera
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Kyung Min Jung
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Heather Barreda
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Carol Paleti
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| | - Joo Youn Oh
- Department of OphthalmologySeoul National University College of MedicineSeoulSouth Korea
| | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, Institute for Regenerative MedicineTexas A&M University School of MedicineCollege StationTexasUSA
| |
Collapse
|
45
|
Lyu Z, Xin M, Oyston DR, Xue T, Kang H, Wang X, Wang Z, Li Q. Cause and consequence of heterogeneity in human mesenchymal stem cells: Challenges in clinical application. Pathol Res Pract 2024; 260:155354. [PMID: 38870711 DOI: 10.1016/j.prp.2024.155354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
Human mesenchymal stem cells (hMSCs) are mesoderm-derived adult stem cells with self-proliferation capacity, pluripotent differentiation potency, and excellent histocompatibility. These advantages make hMSCs a promising tool in clinical application. However, the majority of clinical trials using hMSC therapy for diverse human diseases do not achieve expectations, despite the prospective pre-clinical outcomes in animal models. This is partly attributable to the intrinsic heterogeneity of hMSCs. In this review, the cause of heterogeneity in hMSCs is systematically discussed at multiple levels, including isolation methods, cultural conditions, donor-to-donor variation, tissue sources, intra-tissue subpopulations, etc. Additionally, the effect of hMSCs heterogeneity on the contrary role in tumor progression and immunomodulation is also discussed. The attempts to understand the cellular heterogeneity of hMSCs and its consequences are important in supporting and improving therapeutic strategies for hMSCs.
Collapse
Affiliation(s)
- Zhao Lyu
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Miaomiao Xin
- Assisted Reproductive Center, Women's & Children's Hospital of Northwest, Xi'an, Shaanxi, China; University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, Vodnany, Czech Republic
| | - Dale Reece Oyston
- Department of Evolution, Ecology and Behaviour, University of Liverpool, Liverpool, UK
| | - Tingyu Xue
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Hong Kang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Xiangling Wang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Zheng Wang
- Medical Center of Hematology, the Second Affiliated Hospital, Army Medical University, Chongqing, Sichuan, China.
| | - Qian Li
- Changsha Medical University, Changsha, Hunan, China.
| |
Collapse
|
46
|
Zhan Y, Qian A, Gao J, Ma S, Deng P, Yang H, Zhang X, Li J. Enhancing clinical safety in bioengineered-root regeneration: The use of animal component-free medium. Heliyon 2024; 10:e34173. [PMID: 39092243 PMCID: PMC11292241 DOI: 10.1016/j.heliyon.2024.e34173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
Background Most studies used animal serum-containing medium for bioengineered-root regeneration, but ethical and safety issues raised by animal serum are a potentially significant risk for clinical use. Thus, this study aimed to find a safer method for bioengineered-root regeneration. Methods The biological properties of human dental pulp stem cells (hDPSCs) cultured in animal component-free (ACF) medium or serum-containing medium (5%, 10% serum-containing medium, SCM) were compared in vitro. hDPSCs were cultured in a three-dimensional (3D) environment with human-treated dentin matrix (hTDM). The capacity for odontogenesis was compared using quantitative real-time PCR (qPCR) and Western blot. Subsequently, the hDPSCs/hTDM complexes were transplanted into nude mice subcutaneously. Histological staining was then used to verify the regeneration effect in vivo. Results ACF medium promoted the migration of hDPSCs, but slightly inhibited the proliferation of hDPSCs in the first three days of culture compared to SCM. However, it had no significant effect on cell aging and apoptosis. After 7 days of 3D culture in ACF medium with hTDM, qPCR showed that DMP1, DSPP, OCN, RUNX2, and β-tubulin III were highly expressed in hDPSCs. In addition, 3D cultured hDPSCs/hTDM complexes in ACF medium regenerated dentin, pulp, and periodontal ligament-like tissues similar to SCM groups in vivo. Conclusion ACF medium was proved to be an alternative medium for bioengineered-root regeneration. The strategy of using ACF medium to regenerate bioengineered-root can improve clinical safety for tooth tissue engineering.
Collapse
Affiliation(s)
- Yuzhen Zhan
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Aizhuo Qian
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Jieya Gao
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, The Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Shiyong Ma
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, The Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Pingmeng Deng
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China
| | - Xiaonan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
47
|
Zhang X, Liu T, Ran C, Wang W, Piao F, Yang J, Tian S, Li L, Zhao D. Immunoregulatory paracrine effect of mesenchymal stem cells and mechanism in the treatment of osteoarthritis. Front Cell Dev Biol 2024; 12:1411507. [PMID: 39129785 PMCID: PMC11310049 DOI: 10.3389/fcell.2024.1411507] [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: 04/03/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease caused by chronic inflammation that damages articular cartilage. At present, the treatment of OA includes drug therapy to relieve symptoms and joint replacement therapy for advanced OA. However, these palliatives cannot truly block the progression of the disease from the immunological pathogenesis of OA. In recent years, bone marrow mesenchymal stem cell (BMSC) transplantation has shown great potential in tissue engineering repair. In addition, many studies have shown that BMSC paracrine signals play an important role in the treatment of OA through immune regulation and suppressing inflammation. At present, the mechanism of inflammation-induced OA and the use of BMSC transplantation in joint repair have been reviewed, but the mechanism and significance of BMSC paracrine signals in the treatment of OA have not been fully reviewed. Therefore, this article focused on the latest research progress on the paracrine effects of BMSCs in the treatment of OA and the related mechanisms by which BMSCs secrete cytokines to inhibit the inflammatory response, regulate immune balance, and promote cell proliferation and differentiation. In addition, the application potential of BMSC-Exos as a new type of cell-free therapy for OA is described. This review aimed to provide systematic theoretical support for the clinical application of BMSC transplantation in the treatment of OA.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Tianhao Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Chunxiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Weidan Wang
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Fengyuan Piao
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Jiahui Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Simiao Tian
- Orthopaedic Medical Research Center, Dalian University, Dalian, Liaoning, China
| | - Lu Li
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dewei Zhao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
48
|
Semrau S. Neural Network-Based Filter Design for Compressive Raman Classification of Cells. J Chem Inf Model 2024; 64:5402-5412. [PMID: 38959402 PMCID: PMC11267571 DOI: 10.1021/acs.jcim.3c01856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Cell-based therapies are bound to revolutionize medicine, but significant technical hurdles must be overcome before wider adoption. In particular, nondestructive, label-free methods to characterize cells in real time are needed to optimize the production process and improve quality control. Raman spectroscopy, which provides a fingerprint of a cell's chemical composition, would be an ideal modality but is too slow for high-throughput applications. Compressive Raman techniques, which measure only linear combinations of Raman intensities, can be fast but require careful optimization to deliver high performance. Here, we develop a neural network model to identify optimal parameters for a compressive sensing scheme that reduces measurement time by 2 orders of magnitude. In a data set containing Raman spectra of three different cell types, it achieves up to 90% classification accuracy using only five linear combinations of Raman intensities. Our method thus unlocks the power of Raman spectroscopy for the characterization of cell products.
Collapse
|
49
|
Chen B, Zhu Q, Duan M, Li Q, Wang G, Guan X, Yu P, Xu X, He Y, Xu Y. Optimal Treatment Parameters for Ultrasound-Stimulated Microbubbles in Upregulating Proliferation and Stemness of Bone Marrow Mesenchymal Stem Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1333-1342. [PMID: 38563453 DOI: 10.1002/jum.16457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/16/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Ultrasound-targeted microbubble disruption (UTMD) is a widely used technique to improve the differentiation and proliferation capacity of mesenchymal stem cells (MSCs), but the optimal therapeutic parameters for UTMD are unclear. In this study, we aimed to find the appropriate peak negative pressure (PNP), which is a key parameter for enhancing the stemness properties and proliferation of MSCs. METHODS Experiments were performed in UTMD group, ultrasound (US) group under different PNP exposure conditions (0.5, 1.0, and 1.5 MPa), and control group. Apoptosis safety was analyzed by flow cytometry and MSC proliferation was measured at 12, 24, and 36 hours after irradiation by cell counting kit 8. The expression of the stemness genes NANOG, OCT-4, and SOX-2 were determined by enzyme-linked immunosorbent assay (ELISA) or reverse transcription polymerase chain reaction. RESULTS The results showed that the 1.5 MPa UTMD-treated group had the highest proliferation capacity of MSCs at 24 hours. ELISA or quantitative reverse transcription polymerase chain reaction results showed that UTMD treatment of the 1.5 MPa group significantly upregulated the expression of the stemness genes NANOG, SOX-2, and OCT-4. CONCLUSIONS In conclusion, the appropriate peak PNP value of UTMD was 1.5 MPa, and 1.5 MPa-mediated UTMD group obviously promoted MSCs proliferation and maintained stemness by upregulating the expression of stemness genes.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
- Department of Ultrasound, 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, Shigatse, China
| | - Mao Duan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Qinglong Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Gong Wang
- Department of Ultrasound, Postgraduate Training Basement of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Xue Guan
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Pu Yu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoxun Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ying He
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
50
|
Yi N, Zeng Q, Zheng C, Li S, Lv B, Wang C, Li C, Jiang W, Liu Y, Yang Y, Yan T, Xue J, Xue Z. Functional variation among mesenchymal stem cells derived from different tissue sources. PeerJ 2024; 12:e17616. [PMID: 38952966 PMCID: PMC11216188 DOI: 10.7717/peerj.17616] [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: 01/19/2024] [Accepted: 05/31/2024] [Indexed: 07/03/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) are increasingly recognized for their regenerative potential. However, their clinical application is hindered by their inherent variability, which is influenced by various factors, such as the tissue source, culture conditions, and passage number. Methods MSCs were sourced from clinically relevant tissues, including adipose tissue-derived MSCs (ADMSCs, n = 2), chorionic villi-derived MSCs (CMMSCs, n = 2), amniotic membrane-derived MSCs (AMMSCs, n = 3), and umbilical cord-derived MSCs (UCMSCs, n = 3). Passages included the umbilical cord at P0 (UCMSCP0, n = 2), P3 (UCMSCP3, n = 2), and P5 (UCMSCP5, n = 2) as well as the umbilical cord at P5 cultured under low-oxygen conditions (UCMSCP5L, n = 2). Results We observed that MSCs from different tissue origins clustered into six distinct functional subpopulations, each with varying proportions. Notably, ADMSCs exhibited a higher proportion of subpopulations associated with vascular regeneration, suggesting that they are beneficial for applications in vascular regeneration. Additionally, CMMSCs had a high proportion of subpopulations associated with reproductive processes. UCMSCP5 and UCMSCP5L had higher proportions of subpopulations related to female reproductive function than those for earlier passages. Furthermore, UCMSCP5L, cultured under low-oxygen (hypoxic) conditions, had a high proportion of subpopulations associated with pro-angiogenic characteristics, with implications for optimizing vascular regeneration. Conclusions This study revealed variation in the distribution of MSC subpopulations among different tissue sources, passages, and culture conditions, including differences in functions related to vascular and reproductive system regeneration. These findings hold promise for personalized regenerative medicine and may lead to more effective clinical treatments across a spectrum of medical conditions.
Collapse
Affiliation(s)
- Ning Yi
- Translational Center for Stem Cell Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Stem Cell Research Center, School of Medicine, Tongji University, Shanghai, China
- Hunan Jiahui Genetics Hospital, Changsha, China
| | - Qiao Zeng
- Hunan Jiahui Genetics Hospital, Changsha, China
| | - Chunbing Zheng
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Shiping Li
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Bo Lv
- Translational Center for Stem Cell Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Stem Cell Research Center, School of Medicine, Tongji University, Shanghai, China
- Hunan Jiahui Genetics Hospital, Changsha, China
| | - Cheng Wang
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Chanyi Li
- Translational Center for Stem Cell Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Stem Cell Research Center, School of Medicine, Tongji University, Shanghai, China
| | - Wenjiao Jiang
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Yun Liu
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Yuan Yang
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Tenglong Yan
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| | - Jinfeng Xue
- Translational Center for Stem Cell Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Stem Cell Research Center, School of Medicine, Tongji University, Shanghai, China
- Hunan Jiahui Genetics Hospital, Changsha, China
| | - Zhigang Xue
- Translational Center for Stem Cell Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Stem Cell Research Center, School of Medicine, Tongji University, Shanghai, China
- Hunan Jiahui Genetics Hospital, Changsha, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Yuanpin Cell Technology Co. Ltd., Changsha, China
| |
Collapse
|