1
|
Yao Y, Cao J, Ding C. Mesenchymal stem cells improve osteoarthritis by secreting superoxide dismutase to regulate oxidative stress response. BMC Musculoskelet Disord 2025; 26:409. [PMID: 40275189 DOI: 10.1186/s12891-025-08670-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND To investigate the therapeutic effect of intraarticular injection of mesenchymal stem cells (MSC) in a rabbit osteoarthritis (OA) model. And to suppose whether MSC play a pivotal role in OA therapy by improving oxidative stress through secreting superoxide dismutase (SOD). METHODS MSC and chondrocytes were isolated and cultured in vitro. SOD gene of MSC was silenced by siRNA technology to prepare the SOD-siRNA-MSC for in-vivo study. Twenty healthy adult New Zealand white rabbits underwent papain injection to induce OA and then received intra-articular injection with MSC, siRNA-MSC, or normal saline. The rabbits were divided into 4 groups (n = 5), such as the control group, the model group, the MSC group, the siRNA-MSC group. Cytokines determination was performed 2 and 4 weeks after treatment. Magnetic resonance imaging (MRI) and histopathology and immunohistochemistry determination were performed 4 weeks after treatment. Normal chondrocytes, OA chondrocytes, OA chondrocytes + MSC group, and OA chondrocytes + siRNA-MSC were incubated for 24 h. Then β-galactosidase staining and reactive oxygen species (ROS) level was detected to establish the senescence of the cells. RESULTS COMP, TNF-α, MMP-2 and MMP-13 in the MSC group were significantly decreased compared to those in model group (P < 0.05). However, MMP2 and MMP13 in the siRNA-MSC group were not significantly decreased compared to the model group (P < 0.05). Magnetic resonance results revealed a significant improvement in cartilage and synovial membrane 4 weeks after MSC injection. Histopathology determination showed that cartilage structure was also significantly improved in MSC group. Immunohistochemical analysis revealed amelioration in the expression levels of proteoglycan, COL-2, P21 and P53 in MSC group. On the other hand, MRI, histopathologic and immunohistochemical analysis also indicated a decreased therapeutic effect with SOD-siRNA -MSC. The positive rate of β-galactosidase staining and ROS level of OA chondrocytes were significantly higher than those in normal chondrocytes, which was decreased in OA chondrocytes + MSC (P < 0.05). In addition, it was increased in OA chondrocytes + siRNA-MSC (P < 0.05). CONCLUSION Our study demonstrated for the first time that MSC might be a promising therapy in OA through anti-apoptosis and regeneration in chondrocyte by secreting SOD and improving oxidative stress.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Juan Cao
- Department of Geriatrics, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Congzhu Ding
- Department of Geriatrics, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Weeratunga P, Harman RM, Jager MC, Van de Walle GR. Footprint-free induced pluripotent stem cells can be successfully differentiated into mesenchymal stromal cells in the feline model. Stem Cell Res Ther 2025; 16:195. [PMID: 40254569 PMCID: PMC12010622 DOI: 10.1186/s13287-025-04325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) can propagate indefinitely and give rise to every other cell type, rendering them invaluable for disease modelling, drug development research, and usage in regenerative medicine. While feline iPSCs have been described, there are currently no reports on generating genome integration (footprint)-free iPSCs from domestic cats. Therefore, the objective of this study was to generate feline iPSCs from fetal fibroblasts using non-integrative Sendai virus (SeV) vectors carrying human transcription factors. Moreover, these iPSCs were differentiated into mesenchymal stromal cells (MSCs), which can be used as an alternative to tissue-derived MSCs. METHODS Feline fetal fibroblasts were transduced with CytoTune-iPS 2.0 Sendai Reprogramming vectors at recommended multiplicity of infections (MOI) and cultured for about 6 days. At 7 days post transduction cells were dissociated, replated on inactivated feeder cells and maintained in iPSC medium for 28 days with daily medium change. Emerging iPSC colonies were mechanically passaged and transferred to fresh feeder cells and further passaged every 6-8 days. Four feline iPSC lines were generated, with two selected for further in-depth characterization. Feline iPSCs were then differentiated into MSCs using a serial plating strategy and an inhibitor of the transforming growth factor-β (TGF-β) type I receptor. RESULTS Feline iPSCs exhibited characteristic colony morphology, high nuclear-to-cytoplasmic ratio, positive alkaline phosphatase activity, and expressed feline OCT4, SOX2, and Nanog homeobox (NANOG) stem cell markers. Expression of SeV-derived transgenes decreased during passaging to be eventually lost from the host cells and feline iPSCs could be stably maintained for over 35 passages. Feline iPSCs differentiated into embryoid bodies in vitro and did not form fully differentiated teratomas; instead, they generated in vivo masses containing mesodermal tissue derivatives when injected into immunodeficient mice. Feline iPSC-derived MSCs were plastic adherent, displayed MSC-like morphology, expressed MSC-specific surface markers, and differentiated into cells from the mesodermal lineage in vitro. RNA deep sequencing identified 1,189 differentially expressed genes in feline iPSC-derived MSCs compared to feline iPSCs. CONCLUSION We demonstrated the generation of footprint-free iPSCs from domestic cats and their directed differentiation potential towards MSCs. These SeV-derived feline iPSCs and iPSC-derived MSCs will provide valuable models to study feline diseases and explore novel therapeutic strategies and can serve as translational models for human health, leading to increased knowledge on disease pathogenesis and improved therapeutic interventions.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA
| | - Mason C Jager
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA.
- Department of Veterinary Pathobiology, The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Hosseini N, Kazeminejad E, Oladnabi M, Khosravi A. Isolation and characterization of a new SHED cell line as a standard source for stem cell research and clinical translation. Tissue Cell 2025; 93:102649. [PMID: 39637488 DOI: 10.1016/j.tice.2024.102649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND AIMS Stem cells from human exfoliated deciduous teeth (SHED) are multi-potent mesenchymal stem/stromal cells (MSCs) and are inspected a favorable, non-invasive source beneficial to stem cell-mediated regeneration of damaged tissues. Our aim was to establish and characterize a non-immortalized SHED cell line as an accessible resource and novel platform for stem cell research and tissue regeneration studies. METHODS A Healthy exfoliated deciduous molar was extracted from a 12-year-old girl and shipped to an animal cell culture laboratory. Outgrowing primary cells from explanted small pulp tissues were monitored daily and characterized after passage 3 both morphologically and functionally. The SHED cell line was characterized by calculation of doubling time, cytogenetic analyses, STR analysis, adherence to cell culture flasks under standard cell culture media, and immunophenotypic analysis of specific MSC markers (CD90+, CD73+, CD34- and CD45-) using flow cytometry method. Differentiation potential to osteoblast, adipocyte, and chondrocyte was evaluated under standard differentiation media Expression of OCT-4 and NANOG genes was also assessed using RT-PCR method. RESULTS After the third day, SHED cells were visible. SHED cells were subcultured when they reached 90 % confluence after approximately 17 days. The doubling time of SHED cells was forty seven hours. SHED immunophenotyping showed the high expression level of CD90 (99.2 %) and CD73 (45.9 %), and approximately no expression of CD34 (0.079 %) and CD45 (0.19 %). The human origin, female gender and chromosomal normality of SHED cells was confirmed by cytogenetic analysis. The STR matching analysis showed that SHED cells are well-identified and authentic. No genetic instability and cross-contamination were observed in SHED cells. CONCLUSIONS This study provides a new SHED cell line with a normal karyotype and all the characteristics of MSCs, which can be used as a favorable model cell line in biomedical research and a promising source for clinical translation.
Collapse
Affiliation(s)
- Niloufar Hosseini
- Department of Medical Biotechnology, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezatolah Kazeminejad
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Dental Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Morteza Oladnabi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Gorgan Congenital Malformations Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ayyoob Khosravi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Molecular Medicine, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
4
|
Kumar A, Ramesh S, Kumar V, Mathews JE, Madhuri V. Human second-trimester fetal liver-derived mesenchymal stromal cells are more effective than adult bone marrow MSCs for their superior growth kinetics, immunomodulatory, and osteogenic potential. Tissue Cell 2025; 95:102859. [PMID: 40101501 DOI: 10.1016/j.tice.2025.102859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are promising candidates for cell therapy. Most of the therapeutic applications have used adult bone marrow MSCs, adipose MSCs and perinatal tissue-derived MSCs. Recent evidence suggests that MSCs from mid-gestational fetal tissues are more primitive, grow faster and are biologically more closely related to embryonic stem cells than other sources of MSCs. However, the expression of pluripotency genes raises the question of whether these genes are safe for clinical application. In this study, we demonstrated that second-trimester fetal liver-derived MSCs lack the expression of pluripotent markers and maintain their proliferative and osteogenic differentiation potential beyond passage 12. Compared to other sources, FL-MSCs exhibit characteristics that are promising for use in skeletal regeneration. METHODS MSCs were isolated from the second-trimester fetal liver and characterized for surface antigen expression, pluripotency marker expression and multilineage differentiation. The growth kinetics, population doubling, and number of colony-forming units were analyzed at the 3rd, 5th, 8th and 10th passages of FLMSCs and compared with those of BMMSCs. The immunomodulatory properties of FLMSCs were analyzed by a T-cell proliferation assay. The osteogenic differentiation potential of FL-MSCs was assessed at passages 3, 5, 8 and 12 and compared with that of BMMSCs. RESULTS We demonstrated that second-trimester fetal liver-derived MSCs exhibited a distinct fibroblast-like spindle-shaped morphology and expressed typical MSC surface antigens. Unlike first-trimester fetal MSCs, second-trimester FL-MSCs did not express pluripotent markers and showed significantly greater self-renewal and proliferative potential at higher passages and a lower apoptotic rate than BM-MSCs. Additionally, the osteogenic differentiation potential of FL-MSCs was 4-6 times greater than that of BM-MSCs at both early and late passages CONCLUSION: Our findings underscore the robust self-renewal and proliferative potential of second-trimester fetal liver-derived MSCs, which notably lack pluripotent markers. The ability of FL-MSCs to sustain osteogenic potential through multiple passages makes them promising candidates for bone tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Ashis Kumar
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India; Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala 695011, India
| | - Sowmya Ramesh
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India
| | - Vignesh Kumar
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India
| | | | - Vrisha Madhuri
- Centre for Stem Cell Research, a unit of inStem, Bengaluru, Christian Medical College, Vellore, India; Department of Paediatric Orthopedics, Christian Medical College, Vellore, India; Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala 695011, India.
| |
Collapse
|
5
|
Mahmod AI, Govindaraju K, Lokanathan Y, Said NABM, Ibrahim B. Exploring the Potential of Stem Cells in Modulating Gut Microbiota and Managing Hypertension. Stem Cells Dev 2025; 34:99-116. [PMID: 39836384 DOI: 10.1089/scd.2024.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Hypertension, commonly known as high blood pressure, is a significant health issue that increases the risk of cardiovascular diseases, stroke, and renal failure. This condition broadly encompasses both primary and secondary forms. Despite extensive research, the underlying mechanisms of systemic arterial hypertension-particularly primary hypertension, which has no identifiable cause and is affected by genetic and lifestyle agents-remain complex and not fully understood. Recent studies indicate that an imbalance in gut microbiota, referred to as dysbiosis, may promote hypertension, affecting blood pressure regulation through metabolites such as short-chain fatty acids and trimethylamine N-oxide. Current antihypertensive medications face limitations, including resistance and adherence issues, highlighting the need for novel therapeutic approaches. Stem cell therapy, an emerging field in regenerative medicine, shows promise in addressing these challenges. Stem cells, with mesenchymal stem cells being a prime example, have regenerative, anti-inflammatory, and immunomodulatory properties. Emerging research indicates that stem cells can modulate gut microbiota, reduce inflammation, and improve vascular health, potentially aiding in blood pressure management. Research has shown the positive impact of stem cells on gut microbiota in various disorders, suggesting their potential therapeutic role in treating hypertension. This review synthesizes the recent studies on the complex interactions between gut microbiota, stem cells, and systemic arterial hypertension. By offering a thorough analysis of the current literature, it highlights key insights, uncovers critical gaps, and identifies emerging trends that will inform and guide future investigations in this rapidly advancing field.
Collapse
Affiliation(s)
- Asma Ismail Mahmod
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Kayatri Govindaraju
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Nur Akmarina B M Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Baharudin Ibrahim
- Department of Clinical Pharmacy and Pharmacy Practices, Faculty of Pharmacy, University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Kavaldzhieva K, Mladenov N, Markova M, Belemezova K. Mesenchymal Stem Cell Secretome: Potential Applications in Human Infertility Caused by Hormonal Imbalance, External Damage, or Immune Factors. Biomedicines 2025; 13:586. [PMID: 40149563 PMCID: PMC11940137 DOI: 10.3390/biomedicines13030586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are a source of a wide range of soluble factors, including different proteins, growth factors, cytokines, chemokines, and DNA and RNA molecules, in addition to numerous secondary metabolites and byproducts of their metabolism. MSC secretome can be formally divided into secretory and vesicular parts, both of which are very important for intercellular communication and are involved in processes such as angiogenesis, proliferation, and immunomodulation. Exosomes are thought to have the same content and function as the MSCs from which they are derived, but they also have a number of advantages over stem cells, including low immunogenicity, unaltered functional activity during freezing and thawing, and a lack of tumor formation. In addition, MSC pre-treatment with various inflammatory factors or hypoxia can alter their secretomes so that it can be modified into a more effective treatment. Paracrine factors secreted by MSCs improve the survival of other cell populations by several mechanisms, including immunomodulatory (mostly anti-inflammatory) activity and anti-apoptotic activity partly based on Hsp27 upregulation. Reproductive medicine is one of the fields in which this cell-free approach has been extensively researched. This review presents the possible applications and challenges of using MSC secretome in the treatment of infertility. MSCs and their secretions have been shown to have beneficial effects in various models of female and male infertility resulting from toxic damage, endocrine disorders, trauma, infectious agents, and autoimmune origin.
Collapse
Affiliation(s)
| | | | | | - Kalina Belemezova
- Department of Biology, Medical Faculty, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.K.); (N.M.); (M.M.)
| |
Collapse
|
7
|
Sadiasa A, Werkmeister JA, Gurung S, Gargett CE. Steps towards the clinical application of endometrial and menstrual fluid mesenchymal stem cells for the treatment of gynecological disorders. Expert Opin Biol Ther 2025:1-23. [PMID: 39925343 DOI: 10.1080/14712598.2025.2465826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The human endometrium is a highly regenerative tissue that contains mesenchymal stem/stromal cells (MSCs). These MSCs are sourced via office-based biopsies and menstrual fluid, providing a less invasive and readily available option for cell-based therapies. This review provides an update on endometrial-derived MSCs as a treatment option for gynecological diseases. AREAS COVERED This narrative review covers the characterization and therapeutic mechanisms of endometrium biopsy-derived MSCs (eMSCs) and menstrual fluid-derived mesenchymal stromal cells (MenSCs), highlighting similarities and differences. It also covers studies of their application in preclinical animal models and in clinical trials as potential cell-based therapies for gynecological diseases. EXPERT OPINION eMSCs and MenSCs from a homologous tissue source have the potential to promote regenerative activity as a treatment for gynecological diseases. Both eMSCs and MenSCs demonstrate therapeutic benefits through their paracrine activity in tissue regeneration, immunomodulation, angiogenesis, and mitigating fibrosis. Further research is essential to establish standardized isolation and characterization protocols, particularly for heterogeneous MenSCs, and to fully understand their mechanisms of action. Implementing SUSD2 magnetic bead sorting for purifying eMSCs from endometrial tissues and menstrual fluid is crucial for their use in future cell-based therapies. Optimization of production, storage, and delivery methods will maximize their therapeutic effectiveness.
Collapse
Affiliation(s)
- Alexander Sadiasa
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Shanti Gurung
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
8
|
Koczkowska M, Kostecka A, Zawrzykraj M, Myszczyński K, Skoniecka A, Deptuła M, Tymińska A, Czerwiec K, Jąkalski M, Zieliński J, Crossman DK, Crowley MR, Cichorek M, Skowron PM, Pikuła M, Piotrowski A. Identifying differentiation markers between dermal fibroblasts and adipose-derived mesenchymal stromal cells (AD-MSCs) in human visceral and subcutaneous tissues using single-cell transcriptomics. Stem Cell Res Ther 2025; 16:64. [PMID: 39934849 DOI: 10.1186/s13287-025-04185-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (AD-MSCs) and fibroblasts are both widely used in regenerative medicine, demonstrating significant potential for personalized cell therapy. A major challenge in their use lies in their high biological similarity, encompassing morphology, differentiation capabilities, and flow cytometric markers, making their distinction difficult. METHODS In our study, we aimed to compare AD-MSCs obtained from two types of adipose tissue, subcutaneous and visceral, alongside skin fibroblasts. Notably, all tissue samples were sourced from the same donors. We analyzed the cells for surface antigens via flow cytometry and conducted single-cell RNA sequencing, followed by verification with quantitative PCR (qPCR). RESULTS Our results revealed phenotypic similarities between the isolated AD-MSCs and dermal fibroblasts, particularly in the expression of markers characteristic of AD-MSCs. However, through in-depth analyses, we identified distinct differences between these cell types. Specifically, we pinpointed 30 genes exhibiting the most significant variations in expression between AD-MSCs and fibroblasts. These genes are associated with biological processes such as tissue remodeling, cell movement, and activation in response to external stimuli. Among them, MMP1, MMP3, S100A4, CXCL1, PI16, IGFBP5, COMP were further validated using qPCR, clearly demonstrating their potential to differentiate between AD-MSCs and fibroblasts. CONCLUSIONS Our scRNA-seq analysis elucidates the transcriptional landscape of AD-MSCs and fibroblasts with unprecedented resolution, highlighting both the population-specific markers and the intrapopulation heterogeneity. Our findings underscore the importance of employing high-resolution techniques for cell identification.
Collapse
Affiliation(s)
| | - Anna Kostecka
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Kamil Myszczyński
- Centre of Biostatistics and Bioinformatics Analysis, Medical University of Gdansk, Gdansk, Poland
| | - Aneta Skoniecka
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Milena Deptuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Agata Tymińska
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Katarzyna Czerwiec
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Jąkalski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Jacek Zieliński
- Department of Surgical Oncology, Transplant Surgery and General Surgery, Medical University of Gdansk, Gdansk, Poland
| | - David K Crossman
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael R Crowley
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mirosława Cichorek
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Michał Pikuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland.
| | - Arkadiusz Piotrowski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
9
|
Lu W, Allickson J. Mesenchymal stromal cell therapy: Progress to date and future outlook. Mol Ther 2025:S1525-0016(25)00093-0. [PMID: 39916329 DOI: 10.1016/j.ymthe.2025.02.003] [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: 11/07/2024] [Revised: 01/16/2025] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
In clinical trials, mesenchymal stromal/stem cells (MSCs) have consistently demonstrated safety. However, demonstration of efficacy has been inconsistent and many MSC trials have failed to meet their efficacy endpoint. This disappointing reality is reflected by the limited number MSC therapies approved by regulatory agencies, despite the large number of MSC trials registered on clinicaltrials.gov. Notably, there has been a recent approval of an MSC therapy for pediatric graft-vs.-host disease in the United States, marking the first MSC therapy approved by the U.S. Food and Drug Administration. This review provides a background of the history and potential therapeutic value of MSCs, an overview of MSC products with regulatory approval, and a summary of registered MSC trials. It concludes with a discussion on current and ongoing challenges and questions surrounding MSC therapy that remains to be resolved before becoming available for routine clinical use outside of clinical trials.
Collapse
Affiliation(s)
- Wen Lu
- Department of Laboratory Medicine and Pathology, Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, USA.
| | - Julie Allickson
- Department of Laboratory Medicine and Pathology, Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Figueroa-Valdés AI, Luz-Crawford P, Herrera-Luna Y, Georges-Calderón N, García C, Tobar HE, Araya MJ, Matas J, Donoso-Meneses D, de la Fuente C, Cuenca J, Parra E, Lillo F, Varela C, Cádiz MI, Vernal R, Ortloff A, Nardocci G, Castañeda V, Adasme-Vidal C, Kunze-Küllmer M, Hidalgo Y, Espinoza F, Khoury M, Alcayaga-Miranda F. Clinical-grade extracellular vesicles derived from umbilical cord mesenchymal stromal cells: preclinical development and first-in-human intra-articular validation as therapeutics for knee osteoarthritis. J Nanobiotechnology 2025; 23:13. [PMID: 39806427 PMCID: PMC11730155 DOI: 10.1186/s12951-024-03088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Osteoarthritis (OA) is a joint disease characterized by articular cartilage degradation. Persistent low-grade inflammation defines OA pathogenesis, with crucial involvement of pro-inflammatory M1-like macrophages. While mesenchymal stromal cells (MSC) and their small extracellular vesicles (sEV) hold promise for OA treatment, achieving consistent clinical-grade sEV products remains a significant challenge. This study aims to develop fully characterized, reproducible, clinical-grade batches of sEV derived from umbilical cord (UC)-MSC for the treatment of OA while assessing its efficacy and safety. Initially, a standardized, research-grade manufacturing protocol was established to ensure consistent sEV production. UC-MSC-sEV characterization under non-cGMP conditions showed consistent miRNA and protein profiles, suggesting their potential for standardized manufacturing. In vitro studies evaluated the efficacy, safety, and potency of sEV; animal studies confirmed their effectiveness and safety. In vitro, UC-MSC-sEV polarized macrophages to an anti-inflammatory M2b-like phenotype, through STAT1 modulation, indicating their potential to create an anti-inflammatory environment in the affected joints. In silico studies confirmed sEV's immunosuppressive signature through miRNA and proteome analysis. In an OA mouse model, sEV injected intra-articularly (IA) induced hyaline cartilage regeneration, validated by histological and μCT analyses. The unique detection of sEV signals within the knee joint over time highlights its safety profile by confirming the retention of sEV in the joint. The product development of UC-MSC-sEV involved refining, standardizing, and validating processes in compliance with GMP standards. The initial assessment of the safety of the clinical-grade product via IA administration in a first-in-human study showed no adverse effects after a 12 month follow-up period. These results support the progress of this sEV-based therapy in an early-phase clinical trial, the details of which are presented and discussed in this work. This study provides data on using UC-MSC-sEV as local therapy for OA, highlighting their regenerative and anti-inflammatory properties and safety in preclinical and a proof-of-principle clinical application.
Collapse
Affiliation(s)
- Aliosha I Figueroa-Valdés
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Nicolás Georges-Calderón
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Hugo E Tobar
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - José Matas
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Cirugía Ortopédica, Clínica Universidad de los Andes, Santiago, Chile
| | - Darío Donoso-Meneses
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Jimena Cuenca
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Eliseo Parra
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Fernando Lillo
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Cristóbal Varela
- Departmento de Radiología, Clínica Universidad de los Andes, Santiago, Chile
| | - María Ignacia Cádiz
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Rolando Vernal
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Gino Nardocci
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Verónica Castañeda
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Catalina Adasme-Vidal
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maximiliano Kunze-Küllmer
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
- EVast Bio, Miami, FL, USA
| | - Yessia Hidalgo
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Francisco Espinoza
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Reumatología, Clínica Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
- EVast Bio, Miami, FL, USA.
| | - Francisca Alcayaga-Miranda
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
| |
Collapse
|
11
|
Badralmaa Y, Natarajan V. Aberrant Wnt/β-catenin signaling in the mesenchymal stem cells of LZTFL1-depleted mice leads to increased adipogenesis, with implications for obesity. J Biol Chem 2025; 301:108057. [PMID: 39662832 PMCID: PMC11770550 DOI: 10.1016/j.jbc.2024.108057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Obesity is one of the main clinical characteristics associated with the heterogeneous genetic disorder Bardet-Biedl syndrome (BBS). Leucine zipper transcription factor like 1 (LZTFL1) is a member of the BBS gene family. Our work showed that Lztfl1knockout (LZKO) mice display the obesity phenotype as early as 3 months of age. Mesenchymal stem cells (MSCs) are multipotent stem cells that can differentiate into various cell types, including adipocytes. To understand the role of LZTFL1 in adipogenesis, we analyzed MSCs isolated from LZKO mouse compact bones (CB-MSCs). Compared to wildtype (WT), LZKO CB-MSCs had elongated primary cilia with tapered tips and increased levels of peroxisome proliferator-activated receptor γ (PPARγ), a key transcription factor that favors adipogenesis, and nuclear glucocorticoid receptor (GR), a transcription factor involved in Pparg activation. Also, LZKO CB-MSCs had a lower level of total β-catenin, a core factor of the antiadipogenic canonical Wnt/b-catenin signaling pathway involved in limiting the nuclear localization of GR. Interaction between caveolin1 (CAV1) and LRP6, the main receptor for canonical Wnt signaling, is known to be critical for Wnt pathway activation and β-catenin stabilization. Compared to WT cells, LZKO cells had elevated total, cell-surface, and lipid-raft-associated LRP6 and reduced CAV1, strongly indicating alterations in the components of the Wnt-signaling pathway. We show that in the absence of LZTFL1, adipogenesis-restraining Wnt/β-catenin signaling is inhibited, and adipogenesis-favorable factors are stimulated in CB-MSCs, leading to enhanced adipogenesis. Evidence provided here could help in understanding the mechanism and molecular basis of obesity in LZTFL1-defective patients.
Collapse
Affiliation(s)
- Yunden Badralmaa
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
| |
Collapse
|
12
|
Kumar R, Mahajan S, Gupta U, Madan J, Godugu C, Guru SK, Singh PK, Parvatikar P, Maji I. Stem cell therapy as a novel concept to combat CNS disorders. TARGETED THERAPY FOR THE CENTRAL NERVOUS SYSTEM 2025:175-206. [DOI: 10.1016/b978-0-443-23841-3.00009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Atta H, Kassem DH, Kamal MM, Hamdy NM. Harnessing the ubiquitin proteasome system as a key player in stem cell biology. Biofactors 2025; 51:e2157. [PMID: 39843166 DOI: 10.1002/biof.2157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
Intracellular proteins take part in almost every body function; thus, protein homeostasis is of utmost importance. The ubiquitin proteasome system (UPS) has a fundamental role in protein homeostasis. Its main role is to selectively eradicate impaired or misfolded proteins, thus halting any damage that could arise from the accumulation of these malfunctioning proteins. Proteasomes have a critical role in controlling protein homeostasis in all cell types, including stem cells. We will discuss the role of UPS enzymes as well as the 26S proteasome complex in stem cell biology from several angles. First, we shall overview common trends of proteasomal activity and gene expression of different proteasomal subunits and UPS enzymes upon passaging and differentiation of stem cells toward various cell lineages. Second, we shall explore the effect of modulating proteasomal activity in stem cells and navigate through the interrelation between proteasomes' activity and various proteasome-related transcription factors. Third, we will shed light on curated microRNAs and long non-coding RNAs using various bioinformatics tools that might have a possible role in regulating UPS in stem cells and possibly, upon manipulation, can enhance the differentiation process into different lineages and/or delay senescence upon cell passaging. This will help to decipher the role played by individual UPS enzymes and subunits as well as various interrelated molecular mediators in stem cells' maintenance and/or differentiation and open new avenues in stem cell research. This can ultimately provide a leap toward developing novel therapeutic interventions related to proteasome dysregulation.
Collapse
Affiliation(s)
- Hind Atta
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Drug Research and Development Group, Health Research Center of Excellence, The British University in Egypt, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
14
|
Im GI. Clinical updates in mesenchymal stromal cell therapy for osteoarthritis treatment. Expert Opin Biol Ther 2025; 25:187-195. [PMID: 39710894 DOI: 10.1080/14712598.2024.2446612] [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: 07/26/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) is a common chronic musculoskeletal disease with heterogeneous clinical manifestations and variable responses to different treatments. Unfortunately, there is no effective disease modifying therapy at present that can alter the natural course of the disease. Cell therapy based on mesenchymal stromal cells (MSCs) may offer an attractive therapeutic option for OA with their multiple modes of action, particularly immune-regulatory and regenerative capacities. AREAS COVERED In this narrative review, updates on mode of action based on patient's data, factors that can influence the efficacy of MSC treatment, current status in clinical application of MSCs as seen from randomized, controlled OA trials are introduced as well as the author's perspectives in the future of MSCs as OA therapeutics. EXPERT OPINION Symptomatic relief is not sufficient to justify the high cost associated with culture-expanded stem cells. Its advantages and efficacy over simple and low risk/cost modalities should be seriously reevaluated. Also, as the short-term strategy, efforts should be made to lower the cost of MSC therapy. In the future, multiomics technology may help to predict that subgroup of patients who will favorably respond to stem cell treatment, which would enhance the cost effectiveness and therapeutic benefit of MSC therapy.
Collapse
Affiliation(s)
- Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
15
|
Roato I, Visca M, Mussano F. Suppressing the Aging Phenotype of Mesenchymal Stromal Cells: Are We Ready for Clinical Translation? Biomedicines 2024; 12:2811. [PMID: 39767719 PMCID: PMC11673080 DOI: 10.3390/biomedicines12122811] [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: 10/02/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are involved in the maintenance and regeneration of a large variety of tissues due to their stemness and multi-lineage differentiation capability. Harnessing these advantageous features, a flurry of clinical trials have focused on MSCs to treat different pathologies, but only few protocols have received regulatory approval so far. Among the various causes hindering MSCs' efficacy is the emergence of cellular senescence, which has been correlated with specific characteristics, such as morphological and epigenetic alterations, DNA damage, ROS production, mitochondrial dysfunction, telomere shortening, non-coding RNAs, loss of proteostasis, and a peculiar senescence-associated secretory phenotype. Several strategies have been investigated for delaying or even hopefully reverting the onset of senescence, as assessed by the senescent phenotype of MSCs. Here, the authors reviewed the most updated literature on the potential causes of senescence, with a particular emphasis on the current and future therapeutic approaches aimed at reverting senescence and/or extending the functional lifespan of stem cells.
Collapse
Affiliation(s)
- Ilaria Roato
- Department of Surgical Sciences, CIR-Dental School, University of Turin, 10126 Turin, Italy; (M.V.); (F.M.)
| | | | | |
Collapse
|
16
|
Aswin A, Susilowati H, Yudaniayanti IS, Susanti L, Diyantoro D, Rodprasert W, Kuncorojakti S. Rabbit visceral adipose stromal cell reveals phenotype and genotype characteristics of adult mesenchymal stem cell. Open Vet J 2024; 14:3317-3326. [PMID: 39927335 PMCID: PMC11799644 DOI: 10.5455/ovj.2024.v14.i12.16] [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: 08/22/2024] [Accepted: 11/02/2024] [Indexed: 02/11/2025] Open
Abstract
Background As an excellent model for many animal and human diseases, rabbits are the third-most used mammal model after mice and rats. A plethora of studies on the exploration of rabbit mesenchymal stem cells still face discrepancies, especially in the standardization of phenotype and genotype characteristics to support reproducibility in both biomedical and translational research. Aim This study is aimed to evaluate the characterization and differentiation potential of visceral rabbit adipose-derived mesenchymal stem cells (Rab-ADMSC). Methods Visceral adipose tissue was obtained from three healthy male White New Zealand rabbits. Cells were further processed and cultivated aseptically. Phenotype and genotype assessments, including morphological observation, proliferation capacity, population doubling time, stemness- and senescence-related genes determination, a set panel of mesenchymal stem/stromal cell (MSC) surface markers evaluation, and multilineage differentiation, were performed in this study. Results Visceral Rab-ADMSC exhibited fibroblast-like shape morphology and had a plastic adherent ability, expressed stemness- (NANOG, SOX2) and senescence-related (TP53, CDKN1A) markers. Visceral Rab-ADMSC performs high expression of CD9, moderate expression of CD44 and CD49f, dimly expression of CD105, CD90, and CD73, and negative expression of CD13 and CD133 as well as CD45 as a hematopoietic stem cell marker. Despite these discrepancies, visceral Rab-ADMSC maintained its ability to differentiate into osteocytes, adipocytes, and chondrocytes. Conclusion To recapitulate, visceral Rab-ADMSC reveals the phenotype and genotype characteristics of adult mesenchymal stem cells. The study emphasizes how variations in tissue sources, culture conditions, and techniques can affect the reproducibility and validity of MSC obtained from different specific anatomical depots and species. Thus, the utilization of rabbit MSC as an animal model in biomedical and translational studies should be done with full caution to avoid data misinterpretation.
Collapse
Affiliation(s)
- Ahmad Aswin
- Research Center for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Helen Susilowati
- Research Center for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Ira Sari Yudaniayanti
- Division of Veterinary Clinic, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Lina Susanti
- Division of Veterinary Clinic, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Diyantoro Diyantoro
- Research Center for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Department of Health Science, Faculty of Vocational Studies, Universitas Airlangga, Surabaya, Indonesia
| | - Watchareewan Rodprasert
- Veterinary Stem Cell and Bioengineering Innovation Center, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Suryo Kuncorojakti
- Research Center for Vaccine Technology and Development, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Division of Veterinary Anatomy, Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
17
|
Sharun K, Banu SA, Alifsha B, Abualigah L, Pawde AM, Dhama K, Pal A. Mesenchymal stem cell therapy in veterinary ophthalmology: clinical evidence and prospects. Vet Res Commun 2024; 48:3517-3531. [PMID: 39212813 DOI: 10.1007/s11259-024-10522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mesenchymal stem cell (MSC) therapy presents a promising strategy for treating various ocular conditions in veterinary medicine. This review explores the therapeutic potential of MSCs in managing corneal ulcers, immune-mediated keratitis, chronic superficial keratitis, keratoconjunctivitis sicca, retinal degeneration, and ocular burns in feline, equine, and canine patients. Studies have demonstrated the immunomodulatory and regenerative properties of MSCs, highlighting their ability to mitigate inflammation and promote tissue regeneration. Experimental studies have shown the potential of MSC therapy in reducing corneal opacity and vascularization, indicating significant therapeutic advantages. Delivery methods play a crucial role in optimizing the therapeutic efficacy of MSCs in ocular diseases. Various delivery methods, such as intravitreal injection, subconjunctival injection, topical administration, and scaffold-mediated delivery, are being explored to optimize MSC delivery to the target ocular tissues. Clinical trials have shown significant improvements in clinical signs following MSC therapy, underscoring its efficacy in treating ocular diseases. Additionally, tissue engineering approaches incorporating MSCs, growth factors, and scaffolds offer innovative strategies for corneal regeneration and tissue repair. Despite challenges such as standardization of protocols and long-term safety assessment, ongoing research endeavours seek to unlock the full therapeutic potential of MSC therapy in ocular diseases. Future prospects in MSC therapy involve exploring scaffold and hydrogel-based approaches and cell-free therapies leveraging the bioactive molecules released by MSCs. Continued research and development efforts are essential to unlock the full therapeutic potential of MSCs and realize their transformative impact on ocular diseases in veterinary patients.
Collapse
Affiliation(s)
- Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, 32003, Taiwan.
| | - S Amitha Banu
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - B Alifsha
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laith Abualigah
- Computer Science Department, Al al-Bayt University, Mafraq, 25113, Jordan
- MEU Research Unit, Middle East University, Amman, 11831, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, 11931, Jordan
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
- Artificial Intelligence and Sensing Technologies (AIST) Research Center, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - A M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Amar Pal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
18
|
Chung PH, Lin FH, Liu IH. Enhancing intrinsic TGF-β signaling via heparan sulfate glycosaminoglycan regulation to promote mesenchymal stem cell capabilities and chondrogenesis for cartilage repair. Int J Biol Macromol 2024; 282:137242. [PMID: 39505166 DOI: 10.1016/j.ijbiomac.2024.137242] [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: 09/04/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis burdens patients due to the limited regenerative capacity of chondrocytes. Traditional cartilage repair often falls short, necessitating innovative approaches. Mesenchymal stem cells (MSCs) show promise for regeneration. Heparan sulfate glycosaminoglycans (HS-GAGs) regulate cellular functions, making them a target for cartilage repair. This study highlights how Heparinase III (HepIII) cleaves intact HS-GAGs in bone marrow-derived MSCs (BM-MSCs), enhancing their capabilities and specifically promoting chondrogenesis. HepIII-treated BM-MSCs cultured in a hanging drop device for three days, significantly increased cell number and aggregation into a cell sphere with early chondrogenesis. HepIII promoted BM-MSCs toward chondrogenesis, increasing type II collagen, intact HS-GAGs, and sulfated GAG content, while upregulating chondrogenic and heparan sulfate proteoglycan genes. Treatment with the TGF-β inhibitor (SB-431542) in HepIII-treated BM-MSCs demonstrated enhanced intrinsic transforming growth factor-β (TGF-β) signaling and fibronectin expression. This approach also boosted BM-MSC self-renewal, immunosuppressive potential, and modified acetylated histone signatures, offering a cost-effective strategy for cartilage repair by addressing inflammation, metabolic changes, and the high costs of traditional TGF-β methods. From the results, HepIII-treated BM-MSCs show potential for use in combination with other biopolymers as injectable gels to improve cartilage repair in osteoarthritis patients in the near future.
Collapse
Affiliation(s)
- Pei-Hsuan Chung
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Feng-Huei Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 106, Taiwan; Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli county 350, Taiwan.
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
19
|
Silvana S, Japardi I, Rusda M, Daulay RS, Putra A, Mangunatmadja I, Darlan DM, Sofyani S, Andreas Y. Secretome from hypoxic mesenchymal stem cells as a potential therapy for ischemic stroke: Investigations on VEGF and GFAP expression. NARRA J 2024; 4:e1181. [PMID: 39816066 PMCID: PMC11732002 DOI: 10.52225/narra.v4i2.1181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025]
Abstract
Ischemic stroke is a sudden onset of neurological deficit resulting from a blockage in cerebral blood vessels, which can lead to brain tissue damage, chronic disability, and increased risk of mortality. Secretome from hypoxic mesenchymal stem cells (SH-MSC) is a potential therapy to improve neurological deficit by increasing the expression of vascular endothelial growth factor (VEGF) and reducing glial fibrillary acidic protein (GFAP). These effects can reduce the infarction area of ischemic stroke. Therefore, the aim of this study was to analyze the effect of 150 μL and 300 μL SH-MSC injection on VEGF and GFAP expression as well as the improvement of infarction area in ischemic stroke animal model. A post-test-only experimental design with consecutive sampling was used, with Rattus norvegicus as subjects. Stromal mesenchymal stem cells (S-MSCs) were isolated from the umbilical cords of rats at 21 days of gestation. Secretome production by the S- MSCs was induced under a hypoxic condition, and subsequently isolated. The resultant secretome was administered to rats subjected to middle cerebral artery occlusion (MCAO) at doses of 150 μL (P1 group) and 300 μL (P2 group). The results showed that the infarction area was reduced in P1 (p<0.001) and P2 groups (p<0.001). SH-MSC at a dose of 300 μL increased the expression of VEGF (p=0.028) and reduced the expression of GFAP (p=0.001). In conclusion, secretome from hypoxic S-MSC could potentially improve ischemic stroke by upregulating VEGF expression and downregulating GFAP expression.
Collapse
Affiliation(s)
- Sisca Silvana
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Pediatrics, Faculty of Medicine, Universitas HKBP Nommensen, Medan, Indonesia
| | - Iskandar Japardi
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad Rusda
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Rini S. Daulay
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research (SCCR), Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Indonesia
| | - Irawan Mangunatmadja
- Department of Pediatrics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Dewi M. Darlan
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Sri Sofyani
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Yana Andreas
- Faculty of Medicine, Universitas HKBP Nommensen, Medan, Indonesia
| |
Collapse
|
20
|
Melo WGGD, Bezerra DDO, Silva ERDDFS, Campêlo CB, Carvalho MAMD, Argôlo Neto NM. Behavioral dynamics of medicinal signaling cells from porcine bone marrow in long-term culture. Can J Physiol Pharmacol 2024; 102:672-679. [PMID: 39189463 DOI: 10.1139/cjpp-2023-0458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Medicinal signaling cells (MSC) hold promise for regenerative medicine due to their ability to repair damaged tissues. However, their effectiveness can be affected by how long they are cultured in the lab. This study investigated how passage number influences key properties for regenerative medicine of pig bone marrow MSC. The medicinal signiling cells derived from pig bone marrow (BM-MSC) were cultured in D-MEM High Glucose supplemented with 15% foetal bovine serum until the 25th passage and assessed their growth, viability, ability to differentiate into different cell types (plasticity), and cell cycle activity. Our findings showed that while the cells remained viable until the 25th passage, their ability to grow and differentiate declined after the 5th passage. Additionally, cells in later passages spent more time in a resting phase, suggesting reduced activity. In conclusion, the number of passages is a critical factor for maintaining ideal MSC characteristics. From the 9th passage BM-MSC exhibit decline in proliferation, differentiation potential, and cell cycle activity. Given this, it is possible to suggest that the use of 5th passage cells is the most suitable for therapeutic applications.
Collapse
Affiliation(s)
- Wanderson Gabriel Gomes de Melo
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Dayseanny de Oliveira Bezerra
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | | | - Camile Benício Campêlo
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Maria Acelina Martins de Carvalho
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| | - Napoleão Martins Argôlo Neto
- Núcleo Integrado de Morfologia e Pesquisa com Células-Tronco, Programa de Pós Graduação em Tecnologias Aplicadas a Animais de Interesse Regional, Universidade Federal do Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
21
|
Jiang Y, Harberts J, Assadi A, Chen Y, Spatz JP, Duan W, Nisbet DR, Voelcker NH, Elnathan R. The Roles of Micro- and Nanoscale Materials in Cell-Engineering Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410908. [PMID: 39401098 DOI: 10.1002/adma.202410908] [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: 07/26/2024] [Revised: 09/13/2024] [Indexed: 11/29/2024]
Abstract
Customizable manufacturing of ex vivo cell engineering is driven by the need for innovations in the biomedical field and holds substantial potential for addressing current therapeutic challenges; but it is still only in its infancy. Micro- and nanoscale-engineered materials are increasingly used to control core cell-level functions in cellular engineering. By reprogramming or redirecting targeted cells for extremely precise functions, these advanced materials offer new possibilities. This influences the modularity of cell reprogramming and reengineering, making these materials part of versatile and emerging technologies. Here, the roles of micro- and nanoscale materials in cell engineering are highlighted, demonstrating how they can be adaptively controlled to regulate cellular reprogramming and core cell-level functions, including differentiation, proliferation, adhesion, user-defined gene expression, and epigenetic changes. The current reprogramming routes used to achieve pluripotency from somatic cells and the significant potential of induced pluripotent stem cell technology for translational biomedical research are covered. Recent advances in nonviral intracellular delivery modalities for cell reprogramming and their constraints are evaluated. This paper focuses on emerging physical and combinatorial approaches of intracellular delivery for cell engineering, revealing the capabilities and limitations of these routes. It is showcased how these programmable materials are continually being explored as customizable tools for inducing biophysical stimulation. Harnessing the power of micro- and nanoscale-engineered materials will be a step change in the design of cell engineering, producing a suite of powerful tools for addressing potential future challenges in therapeutic cell engineering.
Collapse
Affiliation(s)
- Yuan Jiang
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Jann Harberts
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Artin Assadi
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Zhejiang, 325000, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Max Planck Schools, 69120, Heidelberg, Germany
| | - Wei Duan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - David R Nisbet
- The Graeme Clark Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, Parkville, VIC, 3010, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Roey Elnathan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
22
|
Pers YM, Soler-Rich R, Vadalà G, Ferreira R, Duflos C, Picot MC, Herman F, Broussous S, Sánchez A, Noriega D, Ardura F, Alberca Zaballos M, García V, Gordillo Cano V, González-Vallinas M, Denaro V, Russo F, Guicheux J, Vilanova J, Orozco L, Meisel HJ, Alfonso M, Rannou F, Maugars Y, Berenbaum F, Barry FP, Tarte K, Louis-Plence P, Ferreira-Dos-Santos G, García-Sancho J, Jorgensen C. Allogenic bone marrow-derived mesenchymal stromal cell-based therapy for patients with chronic low back pain: a prospective, multicentre, randomised placebo controlled trial (RESPINE study). Ann Rheum Dis 2024; 83:1572-1583. [PMID: 39393844 PMCID: PMC11503111 DOI: 10.1136/ard-2024-225771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/10/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES To assess the efficacy of a single intradiscal injection of allogeneic bone marrow mesenchymal stromal cells (BM-MSCs) versus a sham placebo in patients with chronic low back pain (LBP). METHODS Participants were randomised in a prospective, double-blind, controlled study to receive either sham injection or intradiscal injection of 20 million allogeneic BM-MSC, between April 2018 and December 2022. The first co-primary endpoint was the rate of responders defined by improvement of the Visual Analogue Scale (VAS) for pain of at least 20% and 20 mm, or improvement of the Oswestry Disability Index (ODI) of 20% between baseline and month 12. The secondary structural co-primary endpoint was assessed by the disc fluid content measured by quantitative MRI T2, between baseline and month 12. Secondary endpoints included pain VAS, ODI, the Short Form (SF)-36 and the minimal clinically important difference in all timepoints (1, 3, 6, 12 and 24 months). We determined the immune response associated with allogeneic cell injection between baseline and 6 months. Serious adverse events (SAEs) were recorded. RESULTS 114 patients were randomised (n=58, BM-MSC group; n=56, sham placebo group). At 12 months, the primary outcome was not reached (74% in the BM-MSC group vs 69% in the placebo group; p=0.77). The groups did not differ in all secondary outcomes. No SAE related to the intervention occurred. CONCLUSIONS While our study did not conclusively demonstrate the efficacy of allogeneic BM-MSCs for LBP, the procedure was safe. Long-term outcomes of MSC therapy for LBP are still being studied. TRIAL REGISTRATION NUMBER EudraCT 2017-002092-25/ClinicalTrials.gov: NCT03737461.
Collapse
Affiliation(s)
- Yves-Marie Pers
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, CHRU Lapeyronie, Montpellier, France
- INSERM U 1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, France
| | | | - Gianluca Vadalà
- Department of Orthopaedic and Trauma Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - Rosanna Ferreira
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, Montpelliera, France
| | - Claire Duflos
- Clinical Research and Epidemiology Unit (Public Health Department), Montpellier, Languedoc-Roussillon, France
| | - Marie-Christine Picot
- Department of Medical Information, Clinical Research and Epidemiology Unit (Public Health Department), Montpellier, Languedoc-Roussillon, France
| | - Fanchon Herman
- Clinical Research and Epidemiology Unit (Public Health Department), Montpellier, France
| | - Sylvie Broussous
- 7Research and Innovation Department, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Ana Sánchez
- Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), Valladolid, Spain
- Citospin SL, Valladolid, Spain
| | - David Noriega
- Orthopedic Surgery Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Francisco Ardura
- Orthopedic Surgery Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Mercedes Alberca Zaballos
- Citospin SL, Valladolid, Spain
- Department of Biochemistry, Molecular Biology and Physiology, School of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - Verónica García
- Citospin SL, Valladolid, Spain
- Department of Biochemistry, Molecular Biology and Physiology, School of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - Virginia Gordillo Cano
- Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM), Valladolid, Spain
- Citospin SL, Valladolid, Spain
| | - Margarita González-Vallinas
- Citospin SL, Valladolid, Spain
- Department of Biochemistry, Molecular Biology and Physiology, School of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - Vicenzo Denaro
- Department of Orthopaedic and Trauma Surgery, Università Campus Bio-Medico di Roma, Roma, Lazio, Italy
| | - Fabrizio Russo
- Department of Orthopaedic and Trauma Surgery, Università Campus Bio-Medico di Roma, Roma, Lazio, Italy
| | - Jérôme Guicheux
- Regenerative Medicine and Skeleton, RMeS, Nantes Université, Oniris, INSERM, CHU Nantes, Nantes, France
| | | | | | | | - Matias Alfonso
- Orthopaedic Surgery and Traumatology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Yves Maugars
- Regenerative Medicine and Skeleton, RMeS, Nantes Université, Oniris, INSERM, CHU Nantes, Nantes, France
- Service de Rhumatologie, Hôtel-Dieu, CHU Nantes, Nantes, France
| | | | - Frank P Barry
- Regenerative Medicine Institute, University of Galway Regenerative Medicine Institute, Galway, Ireland
| | - Karin Tarte
- Laboratoire SITI, Pôle Biologie, CHU Rennes, Rennes, Bretagne, France
| | - Pascale Louis-Plence
- INSERM U 1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, France
| | - Guilherme Ferreira-Dos-Santos
- Centro Médico Teknon, Barcelona, Spain
- Division of Pain Medicine, Department of Anesthesiology, Reanimation, and Pain Medicine, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Javier García-Sancho
- Citospin SL, Valladolid, Spain
- Department of Biochemistry, Molecular Biology and Physiology, School of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - Christian Jorgensen
- INSERM U 1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France
| |
Collapse
|
23
|
Kangari P, Salahlou R, Vandghanooni S. Harnessing the Therapeutic Potential of Mesenchymal Stem Cells in Cancer Treatment. Adv Pharm Bull 2024; 14:574-590. [PMID: 39494266 PMCID: PMC11530882 DOI: 10.34172/apb.2024.052] [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: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 11/05/2024] Open
Abstract
Cancer, as a complicated disease, is considered to be one of the major leading causes of death globally. Although various cancer therapeutic strategies have been established, however, some issues confine the efficacies of the treatments. In recent decades researchers for finding efficient therapeutic solutions have extensively focused on the abilities of stem cells in cancer inhibition. Mesenchymal stem cells (MSCs) are multipotent stromal cells that can the most widely extracted from various sources such as the bone marrow (BM), placenta, umbilical cord (UC), menses blood, Wharton's jelly (WJ), adipose tissue and dental pulp (DP). These cells are capable of differentiating into the osteoblasts, chondrocytes, and adipocytes. Due to the unique characteristics of MSCs such as paracrine effects, immunomodulation, tumor-tropism, and migration, they are considered promising candidates for cancer therapeutics. Currently, MSCs are an excellent living carrier for delivery of therapeutic genes and chemical agents to target tumor sites. Also, exosomes, the most important extracellular vesicle released from MSCs, act as a strong cell-free tool for cancer therapeutics. MSCs can prevent cancer progression by inhibiting several signaling pathways, such as wnt/β-catenin and PI3K/AKT/mTOR. However, there are several challenges associated with the use of MSCs and their exosomes in the field of therapy that need to be considered. This review explores the significance of MSCs in cell-based therapy, focusing on their homing properties and immunomodulatory characteristics. It also examines the potential of using MSCs as carriers for delivery of anticancer agents and their role in modulating the signal transduction pathways of cancer cells.
Collapse
Affiliation(s)
- Parisa Kangari
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Salahlou
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Lai M, Chen X, Feng J, Ruan Z, Lin J. Morinda officinalis polysaccharide boosts osteogenic differentiation of bone marrow mesenchymal stem cells by Wnt/β-catenin signaling. Am J Transl Res 2024; 16:4492-4503. [PMID: 39398614 PMCID: PMC11470318 DOI: 10.62347/wmli2601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/04/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVES To investigate the role of Morinda officinalis polysaccharide (MOP) in the protein expression of the Wnt/β-catenin signaling cascade during the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), and to elucidate the mechanisms by which MOP enhances osteogenic differentiation at the cellular level. METHODS BMSCs were isolated and cultured using the whole bone marrow adherence method, followed by flow cytometry for the detection of BMSC marker antigens. Two groups were prepared: a low-dose MOP (L-MOP, 10 µg/mL) group and a high-dose MOP (H-MOP, 40 µg/mL) group. MTT assays and cell clone formation assays were performed to evaluate the effects of different MOP doses on BMSC proliferation. Alizarin red staining (ARS) and alkaline phosphatase (ALP) staining were conducted to assess the impact of varying MOP doses on nodule calcification and ALP activity in BMSCs. Additionally, western blot assays were carried out to determine the effects of different MOP concentrations on the expression levels of osteogenesis-related factors and Wnt/β-catenin pathway proteins in BMSCs. RESULTS Highly purified BMSCs were successfully extracted. Subsequent assays demonstrated that BMSCs exhibited enhanced proliferation at all MOP doses, particularly at the H-MOP dose, compared to the control group. Both L-MOP and H-MOP increased calcium content and ALP activity in BMSCs, as well as elevated the expression of osteogenic factors and Wnt/β-catenin pathway proteins compared to the blank control group. However, the addition of Dickkopf-1 (DKK1) significantly reduced BMSC proliferation and osteogenic differentiation compared to the H-MOP group. CONCLUSIONS MOP can enhance BMSC proliferation and osteogenic differentiation by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Manxiang Lai
- Department of Pharmacy, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Xia Chen
- Department of Nursing, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Juan Feng
- Department of Health Management and Biotechnology, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Zhiyan Ruan
- Department of Pharmacy, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Jiwei Lin
- Prevention and Treatment Center, Shenzhen Hospital of Traditional Chinese MedicineShenzhen 518000, Guangdong, China
| |
Collapse
|
25
|
Cyr-Depauw C, Cook DP, Mižik I, Lesage F, Vadivel A, Renesme L, Deng Y, Zhong S, Bardin P, Xu L, Möbius MA, Marzahn J, Freund D, Stewart DJ, Vanderhyden BC, Rüdiger M, Thébaud B. Single-Cell RNA Sequencing Reveals Repair Features of Human Umbilical Cord Mesenchymal Stromal Cells. Am J Respir Crit Care Med 2024; 210:814-827. [PMID: 38564376 DOI: 10.1164/rccm.202310-1975oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/01/2024] [Indexed: 04/04/2024] Open
Abstract
Rationale: The chronic lung disease bronchopulmonary dysplasia (BPD) is the most severe complication of extreme prematurity. BPD results in impaired lung alveolar and vascular development and long-term respiratory morbidity, for which only supportive therapies exist. Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) improve lung structure and function in experimental BPD. Results of clinical trials with MSCs for many disorders do not yet match the promising preclinical studies. A lack of specific criteria to define functionally distinct MSCs persists. Objectives: To determine and correlate single-cell UC-MSC transcriptomic profiles with therapeutic potential. Methods: UC-MSCs from five term donors and human neonatal dermal fibroblasts (HNDFs; control cells of mesenchymal origin) transcriptomes were investigated using single-cell RNA sequencing (scRNA-seq) analysis. The lung-protective effect of UC-MSCs with a distinct transcriptome and control HNDFs was tested in vivo in hyperoxia-induced neonatal lung injury in rats. Measurements and Main Results: UC-MSCs showed limited transcriptomic heterogeneity but were different from HNDFs. Gene Ontology enrichment analysis revealed distinct (progenitor-like and fibroblast-like) UC-MSC subpopulations. Only treatment with progenitor-like UC-MSCs improved lung function and structure and attenuated pulmonary hypertension in hyperoxia-exposed rat pups. Moreover, scRNA-seq identified major histocompatibility complex class I as a molecular marker of nontherapeutic cells and associated with decreased lung retention. Conclusions: UC-MSCs with a progenitor-like transcriptome, but not with a fibroblast-like transcriptome, provide lung protection in experimental BPD. High expression of major histocompatibility complex class I is associated with reduced therapeutic benefit. scRNA-seq may be useful to identify subsets of MSCs with superior repair capacity for clinical application.
Collapse
Affiliation(s)
- Chanèle Cyr-Depauw
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David P Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ivana Mižik
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Flore Lesage
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Laurent Renesme
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Yupu Deng
- Sinclair Centre for Regenerative Medicine and
| | | | - Pauline Bardin
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Liqun Xu
- Sinclair Centre for Regenerative Medicine and
| | - Marius A Möbius
- Neonatology and Pediatric Critical Care Medicine, Department of Pediatrics, University Hospital Carl Gustav Carus, and
- Research Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Jenny Marzahn
- Neonatology and Pediatric Critical Care Medicine, Department of Pediatrics, University Hospital Carl Gustav Carus, and
| | - Daniel Freund
- Research Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Duncan J Stewart
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Obstetrics and Gynecology, University of Ottawa/The Ottawa Hospital, Ottawa, Ontario, Canada; and
| | - Mario Rüdiger
- Neonatology and Pediatric Critical Care Medicine, Department of Pediatrics, University Hospital Carl Gustav Carus, and
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
26
|
Beauregard MA, Bedford GC, Brenner DA, Sanchez Solis LD, Nishiguchi T, Abhimanyu, Longlax SC, Mahata B, Veiseh O, Wenzel PL, DiNardo AR, Hilton IB, Diehl MR. Persistent tailoring of MSC activation through genetic priming. Mol Ther Methods Clin Dev 2024; 32:101316. [PMID: 39282077 PMCID: PMC11396059 DOI: 10.1016/j.omtm.2024.101316] [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: 03/28/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) are an attractive platform for cell therapy due to their safety profile and unique ability to secrete broad arrays of immunomodulatory and regenerative molecules. Yet, MSCs are well known to require preconditioning or priming to boost their therapeutic efficacy. Current priming methods offer limited control over MSC activation, yield transient effects, and often induce the expression of pro-inflammatory effectors that can potentiate immunogenicity. Here, we describe a genetic priming method that can both selectively and sustainably boost MSC potency via the controlled expression of the inflammatory-stimulus-responsive transcription factor interferon response factor 1 (IRF1). MSCs engineered to hyper-express IRF1 recapitulate many core responses that are accessed by biochemical priming using the proinflammatory cytokine interferon-γ (IFN-γ). This includes the upregulation of anti-inflammatory effector molecules and the potentiation of MSC capacities to suppress T cell activation. However, we show that IRF1-mediated genetic priming is much more persistent than biochemical priming and can circumvent IFN-γ-dependent expression of immunogenic MHC class II molecules. Together, the ability to sustainably activate and selectively tailor MSC priming responses creates the possibility of programming MSC activation more comprehensively for therapeutic applications.
Collapse
Affiliation(s)
| | - Guy C. Bedford
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | - Tomoki Nishiguchi
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Abhimanyu
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Santiago Carrero Longlax
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Barun Mahata
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Pamela L. Wenzel
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Andrew R. DiNardo
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Isaac B. Hilton
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Michael R. Diehl
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Chemistry, Rice University, Houston, TX, USA
| |
Collapse
|
27
|
Zhidu S, Ying T, Rui J, Chao Z. Translational potential of mesenchymal stem cells in regenerative therapies for human diseases: challenges and opportunities. Stem Cell Res Ther 2024; 15:266. [PMID: 39183341 PMCID: PMC11346273 DOI: 10.1186/s13287-024-03885-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Advances in stem cell technology offer new possibilities for patients with untreated diseases and disorders. Stem cell-based therapy, which includes multipotent mesenchymal stem cells (MSCs), has recently become important in regenerative therapies. MSCs are multipotent progenitor cells that possess the ability to undergo in vitro self-renewal and differentiate into various mesenchymal lineages. MSCs have demonstrated promise in several areas, such as tissue regeneration, immunological modulation, anti-inflammatory qualities, and wound healing. Additionally, the development of specific guidelines and quality control methods that ultimately result in the therapeutic application of MSCs has been made easier by recent advancements in the study of MSC biology. This review discusses the latest clinical uses of MSCs obtained from the umbilical cord (UC), bone marrow (BM), or adipose tissue (AT) in treating various human diseases such as pulmonary dysfunctions, neurological disorders, endocrine/metabolic diseases, skin burns, cardiovascular conditions, and reproductive disorders. Additionally, this review offers comprehensive information regarding the clinical application of targeted therapies utilizing MSCs. It also presents and examines the concept of MSC tissue origin and its potential impact on the function of MSCs in downstream applications. The ultimate aim of this research is to facilitate translational research into clinical applications in regenerative therapies.
Collapse
Affiliation(s)
- Song Zhidu
- Department of Ophthalmology, the Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun City, Jilin Province, China
| | - Tao Ying
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiang Rui
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhang Chao
- Department of Ophthalmology, the Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun City, Jilin Province, China.
| |
Collapse
|
28
|
Kostecka A, Kalamon N, Skoniecka A, Koczkowska M, Skowron PM, Piotrowski A, Pikuła M. Adipose-derived mesenchymal stromal cells in clinical trials: Insights from single-cell studies. Life Sci 2024; 351:122761. [PMID: 38866216 DOI: 10.1016/j.lfs.2024.122761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Mesenchymal Stromal Cells (MSCs) offer tremendous potential for the treatment of various diseases and their healing properties have been explored in hundreds of clinical trials. These trails primarily focus on immunological and neurological disorders, as well as regenerative medicine. Adipose tissue is a rich source of mesenchymal stromal cells and methods to obtain and culture adipose-derived MSCs (AD-MSCs) have been well established. Promising results from pre-clinical testing of AD-MSCs activity prompted clinical trials that further led to the approval of AD-MSCs for the treatment of complex perianal fistulas in Crohn's disease and subcutaneous tissue defects. However, AD-MSC heterogeneity along with various manufacturing protocols or different strategies to boost their activity create the need for standardized quality control procedures and safety assessment of the intended cell product. High-resolution transcriptomic methods have been recently gaining attention, as they deliver insight into gene expression profiles of individual cells, helping to deconstruct cellular hierarchy and differentiation trajectories, and to understand cell-cell interactions within tissues. This article presents a comprehensive overview of completed clinical trials evaluating the safety and efficacy of AD-MSC treatment, together with current single-cell studies of human AD-MSC. Furthermore, our work emphasizes the increasing significance of single-cell research in elucidating the mechanisms of cellular action and predicting their therapeutic effects.
Collapse
Affiliation(s)
- Anna Kostecka
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Natalia Kalamon
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland.
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Magdalena Koczkowska
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland.
| | - Arkadiusz Piotrowski
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland; 3P - Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, 80-211 Gdańsk, Poland.
| |
Collapse
|
29
|
Moldaschl J, Chariyev-Prinz F, Toegel S, Keck M, Hiden U, Egger D, Kasper C. Spheroid trilineage differentiation model of primary mesenchymal stem/stromal cells under hypoxia and serum-free culture conditions. Front Bioeng Biotechnol 2024; 12:1444363. [PMID: 39144480 PMCID: PMC11321963 DOI: 10.3389/fbioe.2024.1444363] [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: 06/05/2024] [Accepted: 07/12/2024] [Indexed: 08/16/2024] Open
Abstract
Due to their unique properties, human mesenchymal stem/stromal cells (MSCs) possess tremendous potential in regenerative medicine, particularly in cell-based therapies where the multipotency and immunomodulatory characteristics of MSCs can be leveraged to address a variety of disease states. Although MSC-based cell therapeutics have emerged as one of the most promising medical treatments, the clinical translation is hampered by the variability of MSC-based cellular products caused by tissue source-specific differences and the lack of physiological cell culture approaches that closely mimic the human cellular microenvironment. In this study, a model for trilineage differentiation of primary adipose-, bone marrow-, and umbilical cord-derived MSCs into adipocytes, chondrocytes and osteoblasts was established and characterized. Differentiation was performed in spheroid culture, using hypoxic conditions and serum-free and antibiotics-free medium. This platform was characterized for spheroid diameter and trilineage differentiation capacity reflecting functionality of differentiated cells, as indicated by lineage-specific extracellular matrix (ECM) accumulation and expression of distinct secreted markers. The presented model shows spheroid growth during the course of differentiation and successfully supports trilineage differentiation for MSCs from almost all tissue sources except for osteogenesis of umbilical cord-derived MSCs. These findings indicate that this platform provides a suitable and favorable environment for trilineage differentiation of MSCs from various tissue sources. Therefore, it poses a promising model to generate highly relevant biological data urgently required for clinical translation and therefore might be used in the future to generate in vitro microtissues, building blocks for tissue engineering or as disease models.
Collapse
Affiliation(s)
- Julia Moldaschl
- Institute of Cell and Tissue Culture Technologies, BOKU University, Vienna, Austria
| | | | - Stefan Toegel
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Maike Keck
- Department of Plastic, Reconstructive and Aesthetic Surgery, Agaplesion Diakonieklinikum Hamburg, Hamburg, Germany
- Klinik für Plastische Chirurgie, Universität zu Lübeck, Lübeck, Germany
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Dominik Egger
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, BOKU University, Vienna, Austria
| |
Collapse
|
30
|
Bandarra-Tavares H, Franchi-Mendes T, Ulpiano C, Morini S, Kaur N, Harris-Becker A, Vemuri MC, Cabral JMS, Fernandes-Platzgummer A, da Silva CL. Dual production of human mesenchymal stromal cells and derived extracellular vesicles in a dissolvable microcarrier-based stirred culture system. Cytotherapy 2024; 26:749-756. [PMID: 38506771 DOI: 10.1016/j.jcyt.2024.03.001] [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: 07/26/2023] [Revised: 01/29/2024] [Accepted: 03/02/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND & AIMS Cell therapies based on mesenchymal stromal cells (MSCs) have gained an increasing therapeutic interest in the context of multiple disorders. Nonetheless, this field still faces important challenges, particularly concerning suitable manufacturing platforms. Here, we aimed at establishing a scalable culture system to expand umbilical cord-derived Wharton's jelly MSC (MSC(WJ)) and their derived extracellular vesicles (EVs) by using dissolvable microcarriers combined with xeno(geneic)-free culture medium. METHODS MSC(WJ) isolated from three donors were cultured at a starting density of 1 × 106 cells per spinner flask, i.e., 2.8 × 103 cells per cm2 of dissolvable microcarrier surface area. After a 6-day expansion period of MSC(WJ), extracellular vesicles (EVs) were produced for 24 h. RESULTS Taking advantage of an intermittent agitation regimen, we observed high adhesion rates to the microcarriers (over 90% at 24 h) and achieved 15.8 ± 0.7-fold expansion after 6 days of culture. Notably, dissolution of the microcarriers was achieved through a pectinase-based solution to recover the cell product, reducing the hurdles of downstream processing. MSC identity was validated by detecting the characteristic MSC immunophenotype and by multilineage differentiation assays. Considering the growing interest in MSC-derived EVs, which are known to be mediators of the therapeutic features of MSC, this platform also was evaluated for EV production. Upon a 24-h period of conditioning, secreted EVs were isolated by ultrafiltration followed by anion-exchange chromatography and exhibited the typical cup-shaped morphology, small size distribution (162.6 ± 30.2 nm) and expressed EV markers (CD63, CD9 and syntenin-1). CONCLUSIONS Taken together, we established a time-effective and robust scalable platform that complies with clinical-grade standards for the dual production of MSC(WJ) and their derived EV.
Collapse
Affiliation(s)
- Hélder Bandarra-Tavares
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Teresa Franchi-Mendes
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cristiana Ulpiano
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Morini
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Navjot Kaur
- Cell and Gene Therapy, Thermo Fisher Scientific, Cell Biology, Frederick, Maryland, USA
| | - Abigail Harris-Becker
- Cell and Gene Therapy, Thermo Fisher Scientific, Cell Biology, Frederick, Maryland, USA
| | - Mohan C Vemuri
- Cell and Gene Therapy, Thermo Fisher Scientific, Cell Biology, Frederick, Maryland, USA
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
31
|
Nishimaki K, Kaibuchi N, Washio K, Yamato M. Application of mesenchymal stromal cell sheets to prevent medication-related osteonecrosis of the jaw with titanium implants in rats. Odontology 2024; 112:938-949. [PMID: 38367068 DOI: 10.1007/s10266-024-00900-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/04/2024] [Indexed: 02/19/2024]
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is an intractable adverse event. Dental implants are one of the triggering factors of MRONJ, and implant therapy with low MRONJ risk is required. This study aimed to investigate a rat model of MRONJ induced by extraoral placement of titanium materials and the use of mesenchymal stromal cell (MSCs) sheets to prevent MRONJ. Eight-week-old male rats were administered zoledronate and dexamethasone thrice weekly until killing. A week after drug initiation, a titanium screw and a plate were placed on the left buccal side of the mandible. Allogeneic bone marrow-derived MSC sheets were co-grafted with the titanium plates in the MSC sheet ( +) group. Six weeks after titanium placement, the rats were killed, and their excised mandibular bones were subjected to micro-computed tomography (CT) analysis. Histological analysis was performed after the titanium implants were removed. Empty lacunae visualized on hematoxylin and eosin staining were used as evidence of bone necrosis. Bone necrosis was reduced in the MSC sheet ( +) group. Tartrate-resistant acid phosphatase (TRAP) staining revealed a decreased number of TRAP-positive cells in areas with a large number of empty lacunae in the MSC sheet (-) group. Micro-CT analyses demonstrated that the bone volume fraction (BV/TV) was not significantly different between the MSC sheet (-) and ( +) groups. We conclude that MRONJ can be triggered by a titanium placement in rats, and grafting of allogeneic MSC sheets has the potential to prevent MRONJ.
Collapse
Affiliation(s)
- Kazuhiro Nishimaki
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Nobuyuki Kaibuchi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
- Department of Oral and Maxillofacial Surgery, Tokyo Women's Medical University School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Kaoru Washio
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
32
|
Bartolf-Kopp M, Jungst T. The Past, Present, and Future of Tubular Melt Electrowritten Constructs to Mimic Small Diameter Blood Vessels - A Stable Process? Adv Healthc Mater 2024; 13:e2400426. [PMID: 38607966 DOI: 10.1002/adhm.202400426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/20/2024] [Indexed: 04/14/2024]
Abstract
Melt Electrowriting (MEW) is a continuously growing manufacturing platform. Its advantage is the consistent production of micro- to nanometer fibers, that stack intricately, forming complex geometrical shapes. MEW allows tuning of the mechanical properties of constructs via the geometry of deposited fibers. Due to this, MEW can create complex mechanics only seen in multi-material compounds and serve as guiding structures for cellular alignment. The advantage of MEW is also shown in combination with other biotechnological manufacturing methods to create multilayered constructs that increase mechanical approximation to native tissues, biocompatibility, and cellular response. These features make MEW constructs a perfect candidate for small-diameter vascular graft structures. Recently, studies have presented fascinating results in this regard, but is this truly the direction that tubular MEW will follow or are there also other options on the horizon? This perspective will explore the origins and developments of tubular MEW and present its growing importance in the field of artificial small-diameter vascular grafts with mechanical modulation and improved biomimicry and the impact of it in convergence with other manufacturing methods and how future technologies like AI may influence its progress.
Collapse
Affiliation(s)
- Michael Bartolf-Kopp
- Department for Functional Materials in Medicine and Dentistry, Institute of Biofabrication and Functional Materials, University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg, Germany
| | - Tomasz Jungst
- Department for Functional Materials in Medicine and Dentistry, Institute of Biofabrication and Functional Materials, University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg, Germany
- Department of Orthopedics, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
33
|
Barcena AJR, Mishra A, Bolinas DKM, Martin BM, Melancon MP. Integration of Electrospun Scaffolds and Biological Polymers for Enhancing the Delivery and Efficacy of Mesenchymal Stem/Stromal Cell Therapies. FRONT BIOSCI-LANDMRK 2024; 29:228. [PMID: 38940050 PMCID: PMC11725061 DOI: 10.31083/j.fbl2906228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach for a variety of diseases due to their immunomodulatory and tissue regeneration capabilities. Despite their potential, the clinical application of MSC therapies is hindered by limited cell retention and engraftment at the target sites. Electrospun scaffolds, with their high surface area-to-volume ratio and tunable physicochemical properties, can be used as platforms for MSC delivery. However, synthetic polymers often lack the bioactive cues necessary for optimal cell-scaffold interactions. Integrating electrospun scaffolds and biological polymers, such as polysaccharides, proteins, and composites, combines the mechanical integrity of synthetic materials with the bioactivity of natural polymers and represents a strategic approach to enhance cell-scaffold interactions. The molecular interactions between MSCs and blended or functionalized scaffolds have been examined in recent studies, and it has been shown that integration can enhance MSC adhesion, proliferation, and paracrine secretion through the activation of multiple signaling pathways, such as FAK/Src, MAPK, PI3K/Akt, Wnt/β-catenin, and YAP/TAZ. Preclinical studies on small animals also reveal that the integration of electrospun scaffolds and natural polymers represents a promising approach to enhancing the delivery and efficacy of MSCs in the context of regenerating bone, cartilage, muscle, cardiac, vascular, and nervous tissues. Future research should concentrate on identifying the distinct characteristics of the MSC niche, investigating the processes involved in MSC-scaffold interactions, and applying new technologies in stem cell treatment and biofabrication to enhance scaffold design. Research on large animal models and collaboration among materials scientists, engineers, and physicians are crucial to translating these advancements into clinical use.
Collapse
Affiliation(s)
- Allan John R. Barcena
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Archana Mishra
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dominic Karl M. Bolinas
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Benjamin M. Martin
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
34
|
Velikova T, Dekova T, Miteva DG. Controversies regarding transplantation of mesenchymal stem cells. World J Transplant 2024; 14:90554. [PMID: 38947963 PMCID: PMC11212595 DOI: 10.5500/wjt.v14.i2.90554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 04/03/2024] [Indexed: 06/13/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have tantalized regenerative medicine with their therapeutic potential, yet a cloud of controversies looms over their clinical transplantation. This comprehensive review navigates the intricate landscape of MSC controversies, drawing upon 15 years of clinical experience and research. We delve into the fundamental properties of MSCs, exploring their unique immunomodulatory capabilities and surface markers. The heart of our inquiry lies in the controversial applications of MSC transplantation, including the perennial debate between autologous and allogeneic sources, concerns about efficacy, and lingering safety apprehensions. Moreover, we unravel the enigmatic mechanisms surrounding MSC transplantation, such as homing, integration, and the delicate balance between differentiation and paracrine effects. We also assess the current status of clinical trials and the ever-evolving regulatory landscape. As we peer into the future, we examine emerging trends, envisioning personalized medicine and innovative delivery methods. Our review provides a balanced and informed perspective on the controversies, offering readers a clear understanding of the complexities, challenges, and potential solutions in MSC transplantation.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Department of Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Tereza Dekova
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia 1164, Bulgaria
| | | |
Collapse
|
35
|
Kannan S, Gokul Krishna S, Gupta PK, Kolkundkar UK. Advantages of pooling of human bone marrow-derived mesenchymal stromal cells from different donors versus single-donor MSCs. Sci Rep 2024; 14:12654. [PMID: 38825595 PMCID: PMC11144708 DOI: 10.1038/s41598-024-62544-8] [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: 01/05/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024] Open
Abstract
Mesenchymal stromal cells (MSC) from adult bone marrow are the most commonly used cells in clinical trials. MSCs from single donors are the preferred starting material but suffer from a major setback of being heterogeneous that results in unpredictable and inconsistent clinical outcomes. To overcome this, we developed a method of pooling MSCs from different donors and created cell banks to cater clinical needs. Initially, the master cell banks (MCBs) were created at passage 1 (P1) from the bone marrow MSCs isolated from of nine different donors. At this stage, MCBs from three different donors were mixed in equal proportion and expanded till P3 to create working cell banks. Further, the pooled cells and individual donor MSCs were expanded till P5 and cryopreserved and extensively characterised. There was a large heterogeneity among the individual donor MSCs in terms of growth kinetics (90% Coefficient of variation (CV) for cell yield and 44% CV for population doubling time at P5), immunosuppressive ability (30% CV at 1:1 and 300% CV at 1:10 ratio), and the angiogenic factor secretion potential (20% CV for VEGF and71% CV for SDF-1). Comparatively, the pooled cells have more stable profiles (60% CV for cell yield and 7% CV for population doubling time at P5) and exhibit better immunosuppressive ability (15% CV at 1:1 and 32% CV at 1:10 ratio ) and consistent secretion of angiogenic factors (16% CV for VEGF and 51% CV for SDF-1). Further pooling does not compromise the trilineage differentiation capacity or phenotypic marker expression of the MSCs. The senescence and in vitro tumourigenicity characteristics of the pooled cells are also similar to those of individual donor MSCs. We conclude that pooling of MSCs from three different donors reduces heterogeneity among individual donors and produces MSCs with a consistent secretion and higher immunosuppressive profile.
Collapse
Affiliation(s)
- Suresh Kannan
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, ITPL Main Road, Bangalore, Karnataka, 560 048, India.
| | - S Gokul Krishna
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, ITPL Main Road, Bangalore, Karnataka, 560 048, India
| | - Pawan Kumar Gupta
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, ITPL Main Road, Bangalore, Karnataka, 560 048, India
| | - Uday Kumar Kolkundkar
- Stempeutics Research Pvt Ltd, 3rd Floor, Manipal Hospitals Whitefield Pvt. Ltd., #143, EPIP Industrial Area, ITPL Main Road, Bangalore, Karnataka, 560 048, India
| |
Collapse
|
36
|
Matsuzaka Y, Yashiro R. Current Strategies and Therapeutic Applications of Mesenchymal Stem Cell-Based Drug Delivery. Pharmaceuticals (Basel) 2024; 17:707. [PMID: 38931374 PMCID: PMC11206583 DOI: 10.3390/ph17060707] [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: 03/25/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising approach for drug delivery strategies because of their unique properties. These strategies include stem cell membrane-coated nanoparticles, stem cell-derived extracellular vesicles, immunomodulatory effects, stem cell-laden scaffolds, and scaffold-free stem cell sheets. MSCs offer advantages such as low immunogenicity, homing ability, and tumor tropism, making them ideal for targeted drug delivery systems. Stem cell-derived extracellular vesicles have gained attention for their immune properties and tumor-homing abilities, presenting a potential solution for drug delivery challenges. The relationship between MSC-based drug delivery and the self-renewal and differentiation capabilities of MSCs lies in the potential of engineered MSCs to serve as effective carriers for therapeutic agents while maintaining their intrinsic properties. MSCs exhibit potent immunosuppressive functions in MSC-based drug delivery strategies. Stem cell-derived EVs have low immunogenicity and strong therapeutic potential for tissue repair and regeneration. Scaffold-free stem cell sheets represent a cutting-edge approach in regenerative medicine, offering a versatile platform for tissue engineering and regeneration across different medical specialties. MSCs have shown great potential for clinical applications in regenerative medicine because of their ability to differentiate into various cell types, secrete bioactive factors, and modulate immune responses. Researchers are exploring these innovative approaches to enhance drug delivery efficiency and effectiveness in treating various diseases.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan;
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Kiyose 204-8588, Tokyo, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan;
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
37
|
Chu W, Zhang F, Zeng X, He F, Shang G, Guo T, Wang Q, Wu J, Li T, Zhong ZZ, Liang X, Hu J, Liu M. A GMP-compliant manufacturing method for Wharton's jelly-derived mesenchymal stromal cells. Stem Cell Res Ther 2024; 15:131. [PMID: 38702793 PMCID: PMC11069138 DOI: 10.1186/s13287-024-03725-0] [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: 09/05/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) hold great therapeutic potential in regenerative medicine. Therefore, it is crucial to establish a Good Manufacturing Practice (GMP)-compliant methodology for the isolation and culture of WJ-MSCs. Through comprehensive research, encompassing laboratory-scale experiments to pilot-scale studies, we aimed to develop standardized protocols ensuring the high yield and quality of WJ-MSCs manufacturing. METHODS Firstly, optimization of parameters for the enzymatic digestion method used to isolate WJ-MSCs was conducted. These parameters included enzyme concentrations, digestion times, seeding densities, and culture media. Additionally, a comparative analysis between the explant method and the enzymatic digestion method was performed. Subsequently, the consecutive passaging of WJ-MSCs, specifically up to passage 9, was evaluated using the optimized method. Finally, manufacturing processes were developed and scaled up, starting from laboratory-scale flask-based production and progressing to pilot-scale cell factory-based production. Furthermore, a stability study was carried out to assess the storage and use of drug products (DPs). RESULTS The optimal parameters for the enzymatic digestion method were a concentration of 0.4 PZ U/mL Collagenase NB6 and a digestion time of 3 h, resulting in a higher yield of P0 WJ-MSCs. In addition, a positive correlation between the weight of umbilical cord tissue and the quantities of P0 WJ-MSCs has been observed. Evaluation of different concentrations of human platelet lysate revealed that 2% and 5% concentrations resulted in similar levels of cell expansion. Comparative analysis revealed that the enzymatic digestion method exhibited faster outgrowth of WJ-MSCs compared to the explant method during the initial passage. Passages 2 to 5 exhibited higher viability and proliferation ability throughout consecutive passaging. Moreover, scalable manufacturing processes from the laboratory scale to the pilot scale were successfully developed, ensuring the production of high-quality WJ-MSCs. Multiple freeze-thaw cycles of the DPs led to reduced cell viability and viable cell concentration. Subsequent thawing and dilution of the DPs resulted in a significant decrease in both metrics, especially when stored at 20-27 °C. CONCLUSION This study offers valuable insights into optimizing the isolation and culture of WJ-MSCs. Our scalable manufacturing processes facilitate the large-scale production of high-quality WJ-MSCs. These findings contribute to the advancement of WJ-MSCs-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Wanglong Chu
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Fen Zhang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Xiuping Zeng
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Fangtao He
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Guanyan Shang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Tao Guo
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Qingfang Wang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Jianfu Wu
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Tongjing Li
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Zhen Zhong Zhong
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China.
| | - Muyun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, 518000, Shenzhen, Guangdong, People's Republic of China.
- Shenzhen Kenuo Medical Laboratory, 518000, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
38
|
Pignatti E, Maccaferri M, Pisciotta A, Carnevale G, Salvarani C. A comprehensive review on the role of mesenchymal stromal/stem cells in the management of rheumatoid arthritis. Expert Rev Clin Immunol 2024; 20:463-484. [PMID: 38163928 DOI: 10.1080/1744666x.2023.2299729] [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: 09/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease with systemic manifestations. Although the success of immune modulatory drug therapy is considerable, about 40% of patients do not respond to treatment. Mesenchymal stromal/stem cells (MSCs) have been demonstrated to have therapeutic potential for inflammatory diseases. AREAS COVERED This review provides an update on RA disease and on pre-clinical and clinical studies using MSCs from bone marrow, umbilical cord, adipose tissue, and dental pulp, to regulate the immune response. Moreover, the clinical use, safety, limitations, and future perspective of MSCs in RA are discussed. Using the PubMed database and ClincalTrials.gov, peer-reviewed full-text papers, abstracts and clinical trials were identified from 1985 through to April 2023. EXPERT OPINION MSCs demonstrated a satisfactory safety profile and potential for clinical efficacy. However, it is mandatory to deepen the investigations on how MSCs affect the proinflammatory deregulated RA patients' cells. MSCs are potentially good candidates for severe RA patients not responding to conventional therapies but a long-term follow-up after stem cells treatment and standardized protocols are needed. Future research should focus on well-designed multicenter randomized clinical trials with adequate sample sizes and properly selected patients satisfying RA criteria for a valid efficacy evaluation.
Collapse
Affiliation(s)
- Elisa Pignatti
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Monia Maccaferri
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Pisciotta
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Salvarani
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Rheumatology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
39
|
Mikhailov A, Sankai Y. Apoptosis in Postmortal Tissues of Goat Spinal Cords and Survival of Resident Neural Progenitors. Int J Mol Sci 2024; 25:4683. [PMID: 38731901 PMCID: PMC11083117 DOI: 10.3390/ijms25094683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/07/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Growing demand for therapeutic tissue repair recurrently focusses scientists' attention on critical assessment of postmortal collection of live cells, especially stem cells. Our study aimed to assess the survival of neuronal progenitors in postmortal spinal cord and their differentiation potential. Postmortal samples of spinal cords were obtained from human-sized animals (goats) at 6, 12, 24, 36, and 54 h after slaughter. Samples were studied by immunohistology, differentiation assay, Western blot and flow cytometry for the presence and location of GD2-positive neural progenitors and their susceptibility to cell death. TUNEL staining of the goat spinal cord samples over 6-54 h postmortem revealed no difference in the number of positive cells per cross-section. Many TUNEL-positive cells were located in the gray commissure around the central canal of the spinal cord; no increase in TUNEL-positive cells was recorded in either posterior or anterior horns of the gray matter where many GD2-positive neural progenitors can be found. The active caspase 3 amount as measured by Western blot at the same intervals was moderately increasing over time. Neuronal cells were enriched by magnetic separation with antibodies against CD24; among them, the GD2-positive neural progenitor subpopulation did not overlap with apoptotic cells having high pan-caspase activity. Apoptotic cell death events are relatively rare in postmortal spinal cords and are not increased in areas of the neural progenitor cell's location, within measured postmortal intervals, or among the CD24/GD2-positive cells. Data from our study suggest postmortal spinal cords as a valuable source for harvesting highly viable allogenic neural progenitor cells.
Collapse
Affiliation(s)
- Andrey Mikhailov
- Center for Cybernics Research, University of Tsukuba, Tsukuba 305-8573, Japan
| | - Yoshiyuki Sankai
- Faculty of Engineering, Information and Systems, University of Tsukuba, Tsukuba 305-8573, Japan;
| |
Collapse
|
40
|
Tu CC, Zheng ZC, Cheng NC, Yu J, Tai WC, Pan YX, Chang PC. Alveolar mucosal cell spheroids promote extraction socket healing and osseous defect regeneration. J Periodontol 2024; 95:372-383. [PMID: 37531239 DOI: 10.1002/jper.23-0283] [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: 05/02/2023] [Revised: 07/07/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Alveolar mucosa could be a promising source of mesenchymal stem cells (MSCs) for regeneration therapeutics because it exhibits faster healing potential and can be easily collected with minimal periodontal disturbance. This study aimed to evaluate the potential of alveolar mucosal cell (AMC) spheroids for promoting extraction socket healing and calvarial osseous defect regeneration. METHODS AMCs were isolated from Sprague-Dawley rats. Antigenic and MSC surface marker expressions and trilineage differentiation capability were assessed. AMCs were then osteogenically stimulated (OAs) or unstimulated (UAs), self-aggregated to form spheroids, and encapsulated in gelatin hydrogel to fill rat extraction sockets or combined with freeze-dried bone graft (FDBG) to fill rat calvarial osseous defects. The outcome was assessed by gross observation, micro-CT imaging, and immunohistochemistry. RESULTS AMCs highly expressed MSC surface markers, showed weak antigenicity, and were capable of trilineage differentiation at Passage 3. In the extraction sockets, wound closure, socket fill, keratinization, and proliferative activities were accelerated in those with AMC spheroids treatment. Socket fill and maturation were further promoted by OA spheroids. In the calvarial osseous defects, the mineralized tissue ratio was promoted with AMC spheroids/FDBG treatment, and bone sialoprotein expression and cell proliferation were more evident with OA spheroids/FDBG treatment. CONCLUSION AMCs exhibited MSC properties with weak antigenicity. AMC spheroids promoted extraction socket healing, AMC spheroids/FDBG promoted calvarial osseous defect regeneration, and the outcomes were further enhanced by osteogenically stimulation of AMCs.
Collapse
Affiliation(s)
- Che-Chang Tu
- Graduate Institute of Clinical Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Zhao-Cheng Zheng
- Graduate Institute of Clinical Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Nai-Chen Cheng
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Jiashing Yu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Wei-Chiu Tai
- Graduate Institute of Clinical Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Xuan Pan
- Graduate Institute of Clinical Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Chun Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
41
|
Anasiz Y, Ozgul RK, Uckan-Cetinkaya D. Turning Trash Into Treasure: A Simple Protocol for Human Mesenchymal Stromal Cell Isolation From Used Bone Marrow Collection Kits. Curr Protoc 2024; 4:e1032. [PMID: 38606955 DOI: 10.1002/cpz1.1032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The therapeutic potential of mesenchymal stromal cells (MSCs) has been extensively investigated in both preclinical and clinical settings. Recent years have witnessed the emergence of numerous isolation protocols and culture techniques, ranging from the selection of subpopulations to preserve stemness to preconditioning strategies aimed at enhancing therapeutic efficacy, tailored to the specific tissue source. In this protocol, we present a straightforward and cost-effective method for isolating human MSCs (hMSCs) from discarded bone marrow collection kits (comprising bag and filter systems) originally intended for removing impurities and unwanted cellular debris from the collected bone marrow aspirate, ensuring the purity of the stem cell population during stem cell transplantation. Utilizing basic laboratory equipment, we demonstrate the isolation of hMSCs, highlighting the expression of specific surface antigens, and multilineage differentiation into adipogenic, osteogenic, and chondrogenic lineages in vitro. This sustainable and resource-efficient approach not only contributes to reducing medical waste but also holds promise for advancing regenerative medicine applications. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Isolation of human mesenchymal stromal cells from bone marrow collection kits Basic Protocol 2: Culture of human mesenchymal stromal cells Basic Protocol 3: Characterization of human mesenchymal stromal cells with flow cytometry analysis Basic Protocol 4: Characterization of human mesenchymal stromal cells with multilineage differentiation under in vitro conditions.
Collapse
Affiliation(s)
- Yagiz Anasiz
- Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey
- Current affiliation: Department of Medicine, University of Washington, Seattle, Washington
| | - Riza Koksal Ozgul
- Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey
- Department of Pediatric Metabolism, Hacettepe University, Ankara, Turkey
| | - Duygu Uckan-Cetinkaya
- Department of Stem Cell Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University, Ankara, Turkey
- Hematology Unit, Department of Child Health and Diseases, Hacettepe University, Ankara, Turkey
| |
Collapse
|
42
|
Taheri M, Tehrani HA, Dehghani S, Rajabzadeh A, Alibolandi M, Zamani N, Arefian E, Ramezani M. Signaling crosstalk between mesenchymal stem cells and tumor cells: Implications for tumor suppression or progression. Cytokine Growth Factor Rev 2024; 76:30-47. [PMID: 38341337 DOI: 10.1016/j.cytogfr.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stem cells (MSCs) have been extensively used in various therapeutic applications over the last two decades, particularly in regenerative medicine and cancer treatment. MSCs have the ability to differentiate into mesodermal and non-mesodermal lineages, which makes them a popular choice in tissue engineering and regenerative medicine. Studies have shown that MSCs have inherent tumor-suppressive properties and can affect the behavior of multiple cells contributing to tumor development. Additionally, MSCs possess a tumor tropism property and have a hypoimmune nature. The intrinsic features of MSCs along with their potential to undergo genetic manipulation and be loaded with various anticancer therapeutics have motivated researchers to use them in different cancer therapy approaches without considering their complex dynamic biological aspects. However, despite their desirable features, several reports have shown that MSCs possess tumor-supportive properties. These contradictory results signify the sophisticated nature of MSCs and warn against the potential therapeutic applications of MSCs. Therefore, researchers should meticulously consider the biological properties of MSCs in preclinical and clinical studies to avoid any undesirable outcomes. This manuscript reviews preclinical studies on MSCs and cancer from the last two decades, discusses how MSC properties affect tumor progression and explains the mechanisms behind tumor suppressive and supportive functions. It also highlights critical cellular pathways that could be targeted in future studies to improve the safety and effectiveness of MSC-based therapies for cancer treatment. The insights obtained from this study will pave the way for further clinical research on MSCs and development of more effective cancer treatments.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Rajabzadeh
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nina Zamani
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
43
|
Maličev E, Jazbec K. An Overview of Mesenchymal Stem Cell Heterogeneity and Concentration. Pharmaceuticals (Basel) 2024; 17:350. [PMID: 38543135 PMCID: PMC10975472 DOI: 10.3390/ph17030350] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest in cell therapies due to the immunomodulatory and other effects they have after autologous or allogeneic transplantation. In most clinical applications, a high number of MSCs is required; therefore, the isolated MSC population must be expanded in the cell culture until the desired number is reached. Analysing freshly isolated MSCs is challenging due to their rareness and heterogeneity, which is noticeable among donors, tissues, and cell subpopulations. Although the phenotype of MSCs in tissue can differ from those of cultured cells, phenotyping and counting are usually performed only after MSC proliferation. As MSC applicability is a developing and growing field, there is a need to implement phenotyping and counting methods for freshly isolated MSCs, especially in new one-step procedures where isolated cells are implanted immediately without cell culturing. Only by analysing harvested cells can we correctly evaluate such studies. This review describes multilevel heterogeneity and concentrations of MSCs and different strategies for phenotype determination and enumeration of freshly isolated MSCs.
Collapse
Affiliation(s)
- Elvira Maličev
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | - Katerina Jazbec
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
| |
Collapse
|
44
|
Danev N, Li G, Duan J(E, Van de Walle GR. Comparative transcriptomic analysis of bovine mesenchymal stromal cells reveals tissue-source and species-specific differences. iScience 2024; 27:108886. [PMID: 38318381 PMCID: PMC10838956 DOI: 10.1016/j.isci.2024.108886] [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: 09/13/2023] [Revised: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) have the potential to be used as therapeutics, but their efficacy varies due to cellular heterogeneity, which is not fully understood. After characterizing donor-matched bovine MSC from adipose tissue (AT), bone marrow (BM), and peripheral blood (PB), we performed single-cell RNA sequencing (scRNA-seq) to evaluate overarching similarities and differences across these three tissue-derived MSCs. Next, the transcriptomic profiles of the bovine MSCs were compared to those of equine MSCs, derived from the same tissue sources and previously published by our group, and revealed species-specific differences. Finally, the transcriptomic profile from bovine BM-MSCs was compared to mouse and human BM-MSCs and demonstrated that bovine BM-MSCs share more common functionally relevant gene expression profiles with human BM-MSCs than compared to murine BM-MSCs. Collectively, this study presents the cow as a potential non-traditional animal model for translational MSC studies based on transcriptomic profiles similar to human MSCs.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Guangsheng Li
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Jingyue (Ellie) Duan
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
45
|
Mehta JM, Hiremath SC, Chilimba C, Ghasemi A, Weaver JD. Translation of cell therapies to treat autoimmune disorders. Adv Drug Deliv Rev 2024; 205:115161. [PMID: 38142739 PMCID: PMC10843859 DOI: 10.1016/j.addr.2023.115161] [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: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Autoimmune diseases are a diverse and complex set of chronic disorders with a substantial impact on patient quality of life and a significant global healthcare burden. Current approaches to autoimmune disease treatment comprise broadly acting immunosuppressive drugs that lack disease specificity, possess limited efficacy, and confer undesirable side effects. Additionally, there are limited treatments available to restore organs and tissues damaged during the course of autoimmune disease progression. Cell therapies are an emergent area of therapeutics with the potential to address both autoimmune disease immune dysfunction as well as autoimmune disease-damaged tissue and organ systems. In this review, we discuss the pathogenesis of common autoimmune disorders and the state-of-the-art in cell therapy approaches to (1) regenerate or replace autoimmune disease-damaged tissue and (2) eliminate pathological immune responses in autoimmunity. Finally, we discuss critical considerations for the translation of cell products to the clinic.
Collapse
Affiliation(s)
- Jinal M Mehta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Azin Ghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
46
|
dos Santos CC, Lopes-Pacheco M, English K, Rolandsson Enes S, Krasnodembskaya A, Rocco PRM. The MSC-EV-microRNAome: A Perspective on Therapeutic Mechanisms of Action in Sepsis and ARDS. Cells 2024; 13:122. [PMID: 38247814 PMCID: PMC10813908 DOI: 10.3390/cells13020122] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) and MSC-derived extracellular vesicles (EVs) have emerged as innovative therapeutic agents for the treatment of sepsis and acute respiratory distress syndrome (ARDS). Although their potential remains undisputed in pre-clinical models, this has yet to be translated to the clinic. In this review, we focused on the role of microRNAs contained in MSC-derived EVs, the EV microRNAome, and their potential contribution to therapeutic mechanisms of action. The evidence that miRNA transfer in MSC-derived EVs has a role in the overall therapeutic effects is compelling. However, several questions remain regarding how to reconcile the stochiometric issue of the low copy numbers of the miRNAs present in the EV particles, how different miRNAs delivered simultaneously interact with their targets within recipient cells, and the best miRNA or combination of miRNAs to use as therapy, potency markers, and biomarkers of efficacy in the clinic. Here, we offer a molecular genetics and systems biology perspective on the function of EV microRNAs, their contribution to mechanisms of action, and their therapeutic potential.
Collapse
Affiliation(s)
- Claudia C. dos Santos
- Institute of Medical Sciences and Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON M5B 1T8, Canada
- Keenan Center for Biomedical Research, Unity Health Toronto, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22184 Lund, Sweden;
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT9 7BL, UK;
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-599, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro 20020-000, Brazil
| |
Collapse
|
47
|
Kaonis S, Aboellail Z, Forman J, Ghosh S. High-Throughput Multiparametric Quantification of Mechanics Driven Heterogeneity in Mesenchymal Stromal Cell Population. Adv Biol (Weinh) 2024; 8:e2300318. [PMID: 37840408 DOI: 10.1002/adbi.202300318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 10/17/2023]
Abstract
Mesenchymal stromal or stem cells (MSCs) are one of the most promising candidates for a myriad of cell therapy applications. Despite showing promise in numerous preclinical and clinical studies, MSC-based therapy is not yet a reality for regenerative medicine due to its suboptimal outcome at the clinical endpoint. The mechanical environment is a critical determinant of MSC gene expression and function. This study reports that MSC population becomes phenotypically heterogenous and commits to an unwanted osteoprogenitor pathway when it experiences an abnormal mechanically stiff environment, compared to its native softer environment. A method is developed to measure the heterogeneity using nuclear shape, chromatin state, and CD73 marker. Heterogeneity is shown to be associated with a larger spread in the nuclear shape parameters and a smaller spread in the chromatin openness. Subsequently, intervention strategies are investigated to create a more homogeneous MSC population. Culturing MSCs on soft surfaces or inhibiting actomyosin on stiff surfaces can make them more homogeneous, while inhibiting YAP, Runx2, and actin polymerization helps maintain but does not fully homogenize them. This study offers insights for cell and tissue engineers, aiding in the design of optimal conditions and materials for MSC culture, ultimately enhancing their therapeutic potential.
Collapse
Affiliation(s)
- Samantha Kaonis
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
| | - Zack Aboellail
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Jack Forman
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Mechanical Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
- Cell and Molecular Biology, Colorado State University, 1050 Libbie Coy Way, Fort Collins, CO, 80524, USA
| |
Collapse
|
48
|
Chen D. Untoward immune effects of modern medication. J Biomed Res 2023; 38:17-23. [PMID: 38105750 PMCID: PMC10818179 DOI: 10.7555/jbr.37.20230071] [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: 03/30/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 12/19/2023] Open
Abstract
Immune-related adverse events (irAEs) represent an increasingly concerning challenge in the assessment of biopharmaceutical products. In contrast to historically rare allergic reactions associated with small chemical drugs, contemporary biotherapeutics exhibit a significantly higher morbidity of irAEs, because of their complex structure and comprehensive mechanisms of action. While the immunogenicity of protein-based compounds is associated with the induction of anti-drug antibodies, the pathogenesis of irAEs in advanced biologics, such as cell and gene therapy, remains to be further delineated. In the current study, I present an updated profile regarding the untoward immune effects of medications, covering various material categories systematically, with the underlying mechanisms to inspire risk mitigation in biopharmaceutical development and application.
Collapse
Affiliation(s)
- Daohong Chen
- Research Institute, Changshan Biochemical Pharmaceutical, Shijiazhuang, Hebei 050800, China
| |
Collapse
|
49
|
Nakao M, Matsui M, Kim K, Nishiyama N, Grainger DW, Okano T, Kanazawa H, Nagase K. Umbilical cord-derived mesenchymal stem cell sheets transplanted subcutaneously enhance cell retention and survival more than dissociated stem cell injections. Stem Cell Res Ther 2023; 14:352. [PMID: 38072920 PMCID: PMC10712142 DOI: 10.1186/s13287-023-03593-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Human umbilical cord-derived mesenchymal stem cell (hUC-MSC) sheets have recently attracted attention as an alternative approach to injected cell suspensions for stem cell therapy. However, cell engraftment and cytokine expression levels between hUC-MSC sheets and their cell suspensions in vivo have not yet been compared. This study compares hUC-MSC in vivo engraftment efficacy and cytokine expression for both hUC-MSC sheets and cell suspensions. METHODS hUC-MSC sheets were prepared using temperature-responsive cell culture; two types of hUC-MSC suspensions were prepared, either by enzymatic treatment (trypsin) or by enzyme-free temperature reduction using temperature-responsive cell cultureware. hUC-MSC sheets and suspensions were transplanted subcutaneously into ICR mice through subcutaneous surgical placement and intravenous injection, respectively. hUC-MSC sheet engraftment after subcutaneous surgical transplantation was investigated by in vivo imaging while intravenously injected cell suspensions were analyzing using in vitro organ imaging. Cytokine levels in both transplant site tissues and blood were quantified by enzyme-linked immunosorbent assay. RESULTS After subcutaneous transplant, hUC-MSC sheets exhibited longer engraftment duration than hUC-MSC suspensions. This was attributed to extracellular matrix (ECM) and cell-cell junctions retained in sheets but enzymatically altered in suspensions. hUC-MSC suspensions harvested using enzyme-free temperature reduction exhibited relatively long engraftment duration after intravenous injection compared to suspensions prepared using trypsin, as enzyme-free harvest preserved cellular ECM. High HGF and TGF-β1 levels were observed in sheet-transplanted sites compared to hUC-MSC suspension sites. However, no differences in human cytokine levels in murine blood were detected, indicating that hUC-MSC sheets might exert local paracrine rather than endocrine effects. CONCLUSIONS hUC-MSC sheet transplantation could be a more effective cell therapeutic approach due to enhanced engraftment and secretion of therapeutic cytokines over injected hUC-MSC suspensions.
Collapse
Affiliation(s)
- Mitsuyoshi Nakao
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
| | - Hideko Kanazawa
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
50
|
Kim MS, Kong D, Han M, Roh K, Koo H, Lee S, Kang KS. Canine amniotic membrane-derived mesenchymal stem cells ameliorate atopic dermatitis through regeneration and immunomodulation. Vet Res Commun 2023; 47:2055-2070. [PMID: 37421548 DOI: 10.1007/s11259-023-10155-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
Mesenchymal stem cells (MSCs) are a promising tool for treating immune disorders. However, the immunomodulatory effects of canine MSCs compared with other commercialized biologics for treating immune disorders have not been well studied. In this study we investigated the characteristics and immunomodulatory effects of canine amnion membrane (cAM)-MSCs. We examined gene expression of immune modulation and T lymphocytes from activated canine peripheral blood mononuclear cell (PBMC) proliferation. As a result, we confirmed that cAM-MSCs upregulated immune modulation genes (TGF-β1, IDO1 and PTGES2) and suppressed the proliferation capacity of T cells. Moreover, we confirmed the therapeutic effect of cAM-MSCs compared with oclacitinib (OCL), the most commonly used Janus kinase (JAK) inhibitor, as a treatment for canine atopic dermatitis (AD) using a mouse AD model. As a result, we confirmed that cAM-MSCs with PBS treatment groups (passage 4, 6 and 8) compared with PBS only (PBS) though scores of dermatologic signs, tissue pathologic changes and inflammatory cytokines were significantly reduced. In particular, cAM-MSCs were more effective than OCL in the recovery of wound dysfunction, regulation of mast cell activity and expression level of immune modulation protein. Interestingly, subcutaneous injection of cAM-MSCs induced weight recovery, but oral administration of oclacitinib induced weight loss as a side effect. In conclusion, this study suggests that cAM-MSCs can be developed as a safe canine treatment for atopic dermatitis without side effects through effective regeneration and immunomodulation.
Collapse
Affiliation(s)
- Min Soo Kim
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dasom Kong
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Myounghee Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyounghwan Roh
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd, Ace Highend Tower 8, 84, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08590, Republic of Korea
| | - Hojun Koo
- Smile Veterinary Clinic, Jungbu-daero, Cheoin-gu, yongin-si, Gyeonggi-do, 1510, Republic of Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd, Ace Highend Tower 8, 84, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08590, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|