1
|
Hoseini SM, Montazeri F. Cell origin and microenvironment: The players of differentiation capacity in human mesenchymal stem cells. Tissue Cell 2025; 93:102709. [PMID: 39765135 DOI: 10.1016/j.tice.2024.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 03/05/2025]
Abstract
Mesenchymal stem cells (MSCs) have several important properties that make them desirable for regenerative medicine. These properties include immunomodulatory ability, growth factor production, and differentiation into various cell types. Despite extensive research and promising results in clinical trials, our understanding of MSC biology, their mechanism of action, and their targeted and routine use in clinics is limited. Differentiation of human MSCs (hMSCs) is a complex process influenced by various elements such as growth factors, pharmaceutical compounds, microRNAs, 3D scaffolds, and mechanical and electrical stimulation. Research has shown that different culture conditions can affect the differentiation potential of hMSCs obtained from multiple fetal and adult sources. Additionally, it seems that what affects the differentiation capacities of these cells is their secretory characteristics, which are influenced by the origin of the cells and the local microenvironment where the cells are located. The review can provide insights into the microenvironment-based mechanisms involved in MSC differentiation, which can be valuable for future therapeutic applications.
Collapse
Affiliation(s)
- Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| |
Collapse
|
2
|
Zhu S, Liao X, Xu Y, Zhou N, Pan Y, Song J, Zheng T, Zhang L, Bai L, Wang Y, Zhou X, Gou M, Tao J, Liu R. 3D bioprinting of high-performance hydrogel with in-situ birth of stem cell spheroids. Bioact Mater 2025; 43:392-405. [PMID: 39399841 PMCID: PMC11470575 DOI: 10.1016/j.bioactmat.2024.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Digital light processing (DLP)-based bioprinting technology holds immense promise for the advancement of hydrogel constructs in biomedical applications. However, creating high-performance hydrogel constructs with this method is still a challenge, as it requires balancing the physicochemical properties of the matrix while also retaining the cellular activity of the encapsulated cells. Herein, we propose a facile and practical strategy for the 3D bioprinting of high-performance hydrogel constructs through the in-situ birth of stem cell spheroids. The strategy is achieved by loading the cell/dextran microdroplets within gelatin methacryloyl (GelMA) emulsion, where dextran functions as a decoy to capture and aggregate the cells for bioprinting while GelMA enables the mechanical support without losing the structural complexity and fidelity. Post-bioprinting, the leaching of dextran results in a smooth curved surface that promotes in-situ birth of spheroids within hydrogel constructs. This process significant enhances differentiation potential of encapsulated stem cells. As a proof-of-concept, we encapsulate dental pulp stem cells (DPSCs) within hydrogel constructs, showcasing their regenerative capabilities in dentin and neovascular-like structures in vivo. The strategy in our study enables high-performance hydrogel tissue construct fabrication with DLP-based bioprinting, which is anticipated to pave a promising way for diverse biomedical applications.
Collapse
Affiliation(s)
- Shunyao Zhu
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Xueyuan Liao
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Yue Xu
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Nazi Zhou
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Yingzi Pan
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Taijing Zheng
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Lin Zhang
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Liyun Bai
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Yu Wang
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Xia Zhou
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400042, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610065, China
| | - Jie Tao
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Rui Liu
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| |
Collapse
|
3
|
Jiang N, Hu Z, Wang Q, Hao J, Yang R, Jiang J, Wang H. Fibroblast growth factor 2 enhances BMSC stemness through ITGA2-dependent PI3K/AKT pathway activation. J Cell Physiol 2024; 239:e31423. [PMID: 39188080 DOI: 10.1002/jcp.31423] [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: 03/29/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSC) are promising cellular reservoirs for treating degenerative diseases, tissue injuries, and immune system disorders. However, the stemness of BMSCs tends to decrease during in vitro cultivation, thereby restricting their efficacy in clinical applications. Consequently, investigating strategies that bolster the preservation of BMSC stemness and maximize therapeutic potential is necessary. Transcriptomic and single-cell sequencing methodologies were used to perform a comprehensive examination of BMSCs with the objective of substantiating the pivotal involvement of fibroblast growth factor 2 (FGF2) and integrin alpha 2 (ITGA2) in stemness regulation. To investigate the impact of these genes on the BMSC stemness in vitro, experimental approaches involving loss and gain of function were implemented. These approaches encompassed the modulation of FGF2 and ITGA2 expression levels via small interfering RNA and overexpression plasmids. Furthermore, we examined their influence on the proliferation and differentiation capacities of BMSCs, along with the expression of stemness markers, including octamer-binding transcription factor 4, Nanog homeobox, and sex determining region Y-box 2. Transcriptomic analyzes successfully identified FGF2 and ITGA2 as pivotal genes responsible for regulating the stemness of BMSCs. Subsequent single-cell sequencing revealed that elevated FGF2 and ITGA2 expression levels within specific stem cell subpopulations are closely associated with stemness maintenance. Moreover, additional in vitro experiments have convincingly demonstrated that FGF2 effectively enhances the BMSC stemness by upregulating ITGA2 expression, a process mediated by the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. This conclusion was supported by the observed upregulation of stemness markers following the induction of FGF2 and ITGA2. Moreover, administration of the BEZ235 pathway inhibitor resulted in the repression of stemness transcription factors, suggesting the substantial involvement of the PI3K/AKT pathway in stemness preservation facilitated by FGF2 and ITGA2. This study elucidates the involvement of FGF2 in augmenting BMSC stemness by modulating ITGA2 and activating the PI3K/AKT pathway. These findings offer valuable contributions to stem cell biology and emphasize the potential of manipulating FGF2 and ITGA2 to optimize BMSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Nizhou Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
- Department of Spine Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhenxin Hu
- Department of Spine Surgery, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Quanxiang Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jiayu Hao
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Rui Yang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Jian Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Hong Wang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| |
Collapse
|
4
|
Augustine R, Gezek M, Nikolopoulos VK, Buck PL, Bostanci NS, Camci-Unal G. Stem Cells in Bone Tissue Engineering: Progress, Promises and Challenges. Stem Cell Rev Rep 2024; 20:1692-1731. [PMID: 39028416 DOI: 10.1007/s12015-024-10738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/20/2024]
Abstract
Bone defects from accidents, congenital conditions, and age-related diseases significantly impact quality of life. Recent advancements in bone tissue engineering (TE) involve biomaterial scaffolds, patient-derived cells, and bioactive agents, enabling functional bone regeneration. Stem cells, obtained from numerous sources including umbilical cord blood, adipose tissue, bone marrow, and dental pulp, hold immense potential in bone TE. Induced pluripotent stem cells and genetically modified stem cells can also be used. Proper manipulation of physical, chemical, and biological stimulation is crucial for their proliferation, maintenance, and differentiation. Stem cells contribute to osteogenesis, osteoinduction, angiogenesis, and mineralization, essential for bone regeneration. This review provides an overview of the latest developments in stem cell-based TE for repairing and regenerating defective bones.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Radiology, Stanford Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | | | - Paige Lauren Buck
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
5
|
Shao Z, Wang B, Gao H, Zhang S. Microenvironmental interference with intra-articular stem cell regeneration influences the onset and progression of arthritis. Front Genet 2024; 15:1380696. [PMID: 38841721 PMCID: PMC11150611 DOI: 10.3389/fgene.2024.1380696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Studies have indicated that the preservation of joint health and the facilitation of damage recovery are predominantly contingent upon the joint's microenvironment, including cell-cell interactions, the extracellular matrix's composition, and the existence of local growth factors. Mesenchymal stem cells (MSCs), which possess the capacity to self-renew and specialize in many directions, respond to cues from the microenvironment, and aid in the regeneration of bone and cartilage, are crucial to this process. Changes in the microenvironment (such as an increase in inflammatory mediators or the breakdown of the extracellular matrix) in the pathological context of arthritis might interfere with stem cell activation and reduce their ability to regenerate. This paper investigates the potential role of joint microenvironmental variables in promoting or inhibiting the development of arthritis by influencing stem cells' ability to regenerate. The present status of research on stem cell activity in the joint microenvironment is also outlined, and potential directions for developing new treatments for arthritis that make use of these intervention techniques to boost stem cell regenerative potential through altering the intra-articular environment are also investigated. This review's objectives are to investigate these processes, offer fresh perspectives, and offer a solid scientific foundation for the creation of arthritic treatment plans in the future.
Collapse
Affiliation(s)
| | | | | | - Shenqi Zhang
- Department of Joint and Sports Medicine, Zaozhuang Municipal Hospital Affiliated to Jining Medical University, Zaozhuang, Shandong, China
| |
Collapse
|
6
|
Loda A, Semeraro F, Parolini S, Ronca R, Rezzola S. Cancer stem-like cells in uveal melanoma: novel insights and therapeutic implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189104. [PMID: 38701937 DOI: 10.1016/j.bbcan.2024.189104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Uveal melanoma (UM) is the most common primary ocular tumor in the adult population. Even though these primary tumors are successfully treated in 90% of cases, almost 50% of patients ultimately develop metastasis, mainly in the liver, via hematological dissemination, with a median survival spanning from 6 to 12 months after diagnosis. In this context, chemotherapy regimens and molecular targeted therapies have demonstrated poor response rates and failed to improve survival. Among the multiple reasons for therapy failure, the presence of cancer stem-like cells (CSCs) represents the main cause of resistance to anticancer therapies. In the last few years, the existence of CSCs in UM has been demonstrated both in preclinical and clinical studies, and new molecular pathways and mechanisms have been described for this subpopulation of UM cells. Here, we will discuss the state of the art of CSC biology and their potential exploitation as therapeutic target in UM.
Collapse
Affiliation(s)
- Alessandra Loda
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesco Semeraro
- Eye Clinic, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; National Center for Gene Therapy and Drugs based on RNA Technology - CN3, Padova, Italy; Consorzio Interuniversitario per le Biotecnologie (CIB), Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Consorzio Interuniversitario per le Biotecnologie (CIB), Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
7
|
Xue Z, Liao Y, Li Y. Effects of microenvironment and biological behavior on the paracrine function of stem cells. Genes Dis 2024; 11:135-147. [PMID: 37588208 PMCID: PMC10425798 DOI: 10.1016/j.gendis.2023.03.013] [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: 01/08/2023] [Revised: 02/14/2023] [Accepted: 03/05/2023] [Indexed: 08/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs), the most well-studied cell type in the field of stem cell therapy, have multi-lineage differentiation and self-renewal potential. MSC-based therapies have been used to treat diverse diseases because of their ability to potently repair tissue and locally restore function. An increasing body of evidence demonstrates that paracrine function is central to the effects of MSC-based therapy. Growth factors, cytokines, chemokines, extracellular matrix components, and extracellular vehicles all contribute to the beneficial effects of MSCs on tissue regeneration and repair. The paracrine substances secreted by MSCs change depending on the tissue microenvironment and biological behavior. In this review, we discuss the bioactive substances secreted by MSCs depending on the microenvironment and biological behavior and their regulatory mechanisms, which explain their potential to treat human diseases, to provide new ideas for further research and clinical cell-free therapy.
Collapse
Affiliation(s)
- Zhixin Xue
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yunjun Liao
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ye Li
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
8
|
Park S, Kim S, Lim K, Shin Y, Song K, Kang GH, Kim DY, Shin HC, Cho SG. Thermostable Basic Fibroblast Growth Factor Enhances the Production and Activity of Human Wharton's Jelly Mesenchymal Stem Cell-Derived Extracellular Vesicles. Int J Mol Sci 2023; 24:16460. [PMID: 38003648 PMCID: PMC10671285 DOI: 10.3390/ijms242216460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Wharton's jelly-derived mesenchymal stem cell (WJ-MSC)-derived exosomes contain a diverse cargo and exhibit remarkable biological activity, rendering them suitable for regenerative and immune-modulating functions. However, the quantity of secretion is insufficient. A large body of prior work has investigated the use of various growth factors to enhance MSC-derived exosome production. In this study, we evaluated the utilization of thermostable basic fibroblast growth factor (TS-bFGF) with MSC culture and exosome production. MSCs cultured with TS-bFGF displayed superior proliferation, as evidenced by cell cycle analysis, compared with wild-type bFGF (WT-bFGF). Stemness was assessed through mRNA expression level and colony-forming unit (CFU) assays. Furthermore, nanoparticle tracking analysis (NTA) measurements revealed that MSCs cultured with TS-bFGF produced a greater quantity of exosomes, particularly under three-dimensional culture conditions. These produced exosomes demonstrated substantial anti-inflammatory and wound-healing effects, as confirmed by nitric oxide (NO) assays and scratch assays. Taken together, we demonstrate that utilization of TS-bFGF for WJ-MSC-derived exosome production not only increases exosome yield but also enhances the potential for various applications in inflammation regulation and wound healing.
Collapse
Affiliation(s)
- SangRok Park
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
| | - SeJong Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - KyungMin Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - YeoKyung Shin
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kwonwoo Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Geun-Ho Kang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dae Young Kim
- PnP Biopharm Co., Ltd., 1304, Acetechnotower 8-cha, 11 Digital-ro 33-gil, Guro-gu, Seoul 08380, Republic of Korea; (D.Y.K.); (H.-C.S.)
| | - Hang-Cheol Shin
- PnP Biopharm Co., Ltd., 1304, Acetechnotower 8-cha, 11 Digital-ro 33-gil, Guro-gu, Seoul 08380, Republic of Korea; (D.Y.K.); (H.-C.S.)
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.P.); (S.K.); (K.L.); (Y.S.); (K.S.); (G.-H.K.)
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
9
|
Jafar H, Ahmed K, Rayyan R, Sotari S, Buqain R, Ali D, Al Bdour M, Awidi A. Plasma-Treated Electrospun PLGA Nanofiber Scaffold Supports Limbal Stem Cells. Polymers (Basel) 2023; 15:4244. [PMID: 37959924 PMCID: PMC10648479 DOI: 10.3390/polym15214244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
The corneal epithelial layer is continuously replaced by limbal stem cells. Reconstructing this layer in vitro using synthetic scaffolds is highly needed. Poly-lactic-co-glycolic acid (PLGA) is approved for human use due to its biocompatibility and biodegradability. However, PLGA is hydrophobic, preventing cell adherence to PLGA membranes. PLGA scaffolds were prepared by electrospinning on a custom-made target drum spinning at a rate of 1000 rpm with a flow rate of 0.5 mL/h and voltage at 20 kV, then treated with oxygen plasma at 30 mA using a vacuum coater. Scaffolds were characterized by SEM, mechanically by tensile testing, and thermally by DSC and TGA. In vitro degradation was measured by weight loss and pH drop. Wettability was assessed through water uptake and contact angles measurements. Human limbal stem cells (hLSCs) were isolated and seeded on the scaffolds. Cell attachment and cytotoxicity assay were evaluated on day 1 and 5 after cell seeding. SEM showed regular fiber morphology with diameters ranging between 150 nm and 950 nm. Tensile strength demonstrated similar average stress values for both plasma- and non-plasma-treated samples. Scaffolds also showed gradual degradability over a period of 7-8 weeks. Water contact angle and water absorption were significantly enhanced for plasma-treated scaffolds, indicating a favorable increase in their hydrophilicity. Scaffolds have also supported hLSCs growth and attachment with no signs of cytotoxicity. We have characterized a nanofiber electrospun plasma-treated PLGA scaffold to investigate the mechanical and biological properties and the ability to support the attachment and maintenance of hLSCs.
Collapse
Affiliation(s)
- Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (H.J.)
| | - Khalid Ahmed
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Rama Rayyan
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Shorouq Sotari
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (H.J.)
| | - Rula Buqain
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (H.J.)
| | - Dema Ali
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (H.J.)
| | - Muawyah Al Bdour
- Department of Ophthalmology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; (H.J.)
- Thrombosis Homeostasis Laboratory, School of Medicine, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
10
|
Rehman A, Nigam A, Laino L, Russo D, Todisco C, Esposito G, Svolacchia F, Giuzio F, Desiderio V, Ferraro G. Mesenchymal Stem Cells in Soft Tissue Regenerative Medicine: A Comprehensive Review. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1449. [PMID: 37629738 PMCID: PMC10456353 DOI: 10.3390/medicina59081449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023]
Abstract
Soft tissue regeneration holds significant promise for addressing various clinical challenges, ranging from craniofacial and oral tissue defects to blood vessels, muscle, and fibrous tissue regeneration. Mesenchymal stem cells (MSCs) have emerged as a promising tool in regenerative medicine due to their unique characteristics and potential to differentiate into multiple cell lineages. This comprehensive review explores the role of MSCs in different aspects of soft tissue regeneration, including their application in craniofacial and oral soft tissue regeneration, nerve regeneration, blood vessel regeneration, muscle regeneration, and fibrous tissue regeneration. By examining the latest research findings and clinical advancements, this article aims to provide insights into the current state of MSC-based therapies in soft tissue regenerative medicine.
Collapse
Affiliation(s)
- Ayesha Rehman
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (A.R.); (A.N.)
| | - Aditya Nigam
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (A.R.); (A.N.)
| | - Luigi Laino
- Multidisciplinary Department of Medicine for Surgery and Orthodontics, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (L.L.); (D.R.); (G.F.)
| | - Diana Russo
- Multidisciplinary Department of Medicine for Surgery and Orthodontics, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (L.L.); (D.R.); (G.F.)
| | | | | | - Fabiano Svolacchia
- Departments of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00118 Rome, Italy;
| | - Federica Giuzio
- Department of Sciences, University of Basilicata, Via Nazario Sauro 85, 85100 Potenza, Italy;
- U.O.S.D. of Plastic Surgery A.O.R “San Carlo”, 85100 Potenza, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (A.R.); (A.N.)
| | - Giuseppe Ferraro
- Multidisciplinary Department of Medicine for Surgery and Orthodontics, University of Campania “Luigi Vanvitelli”, Via L. Armanni 5, 80138 Naples, Italy; (L.L.); (D.R.); (G.F.)
| |
Collapse
|
11
|
Loda A, Calza S, Giacomini A, Ravelli C, Krishna Chandran AM, Tobia C, Tabellini G, Parolini S, Semeraro F, Ronca R, Rezzola S. FGF-trapping hampers cancer stem-like cells in uveal melanoma. Cancer Cell Int 2023; 23:89. [PMID: 37165394 PMCID: PMC10173517 DOI: 10.1186/s12935-023-02903-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/24/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Cancer stem-like cells (CSCs) are a subpopulation of tumor cells responsible for tumor initiation, metastasis, chemoresistance, and relapse. Recently, CSCs have been identified in Uveal Melanoma (UM), which represents the most common primary tumor of the eye. UM is highly resistant to systemic chemotherapy and effective therapies aimed at improving overall survival of patients are eagerly required. METHODS Herein, taking advantage from a pan Fibroblast Growth Factor (FGF)-trap molecule, we singled out and analyzed a UM-CSC subset with marked stem-like properties. A hierarchical clustering of gene expression data publicly available on The Cancer Genome Atlas (TCGA) was performed to identify patients' clusters. RESULTS By disrupting the FGF/FGF receptor (FGFR)-mediated signaling, we unmasked an FGF-sensitive UM population characterized by increased expression of numerous stemness-related transcription factors, enhanced aldehyde dehydrogenase (ALDH) activity, and tumor-sphere formation capacity. Moreover, FGF inhibition deeply affected UM-CSC survival in vivo in a chorioallantoic membrane (CAM) tumor graft assay, resulting in the reduction of tumor growth. At clinical level, hierarchical clustering of TCGA gene expression data revealed a strong correlation between FGFs/FGFRs and stemness-related genes, allowing the identification of three distinct clusters characterized by different clinical outcomes. CONCLUSIONS Our findings support the evidence that the FGF/FGFR axis represents a master regulator of cancer stemness in primary UM tumors and point to anti-FGF treatments as a novel therapeutic strategy to hit the CSC component in UM.
Collapse
Affiliation(s)
- Alessandra Loda
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Stefano Calza
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Adwaid Manu Krishna Chandran
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Chiara Tobia
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Francesco Semeraro
- Eye Clinic, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy.
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy.
| |
Collapse
|
12
|
Lee K, Jackson A, John N, Zhang R, Ozhava D, Bhatia M, Mao Y. Bovine Fibroblast-Derived Extracellular Matrix Promotes the Growth and Preserves the Stemness of Bovine Stromal Cells during In Vitro Expansion. J Funct Biomater 2023; 14:jfb14040218. [PMID: 37103308 PMCID: PMC10144935 DOI: 10.3390/jfb14040218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
Cultivated meat is a fast-growing research field and an industry with great potential to overcome the limitations of traditional meat production. Cultivated meat utilizes cell culture and tissue engineering technologies to culture a vast number of cells in vitro and grow/assemble them into structures mimicking the muscle tissues of livestock animals. Stem cells with self-renewal and lineage-specific differentiation abilities have been considered one of the key cell sources for cultivated meats. However, the extensive in vitro culturing/expansion of stem cells results in a reduction in their abilities to proliferate and differentiate. Extracellular matrix (ECM) has been used as a culturing substrate to support cell expansion for cell-based therapies in regenerative medicine due to its resemblance to the native microenvironment of cells. In this study, the effect of the ECM on the expansion of bovine umbilical cord stromal cells (BUSC) in vitro was evaluated and characterized. BUSCs with multi-lineage differentiation potentials were isolated from bovine placental tissue. Decellularized ECM prepared from a confluent monolayer of bovine fibroblasts (BF) is free of cellular components but contains major ECM proteins such as fibronectin and type I collagen and ECM-associated growth factors. Expansion of BUSC on ECM for three passages (around three weeks) resulted in about 500-fold amplification, while cells were amplified less than 10-fold when cultured on standard tissue culture plates (TCP). Moreover, the presence of ECM reduced the requirement for serum in the culture medium. Importantly, the cells amplified on ECM retained their differentiation abilities better than cells cultured on TCP. The results of our study support the notion that monolayer cell-derived ECM may be a strategy to expand bovine cells in vitro effectively and efficiently.
Collapse
Affiliation(s)
- Kathleen Lee
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Anisha Jackson
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Nikita John
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Ryan Zhang
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Derya Ozhava
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Mohit Bhatia
- Atelier Meats, 666 Burrard Street, Suite 500, Vancouver, BC V6C 3P6, Canada
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
13
|
Shifa ul Haq H, Ashfaq R, Mehmood A, Shahid W, Azam G, Azam M, Tasneem S, Akram SJ, Malik K, Riazuddin S. Priming with caffeic acid enhances the potential and survival ability of human adipose-derived stem cells to counteract hypoxia. Regen Ther 2023; 22:115-127. [PMID: 36751276 PMCID: PMC9883200 DOI: 10.1016/j.reth.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023] Open
Abstract
The therapeutic effectiveness of stem cells after transplantation is hampered by the hypoxic milieu of chronic wounds. Prior research has established antioxidant priming as a thorough plan to improve stem cell performance. The purpose of this study was to ascertain how caffeic acid (CA) priming affected the ability of human adipose-derived stem cells (hASCs) to function under hypoxic stress. In order to study the cytoprotective properties of CA, hASCs were primed with CA in CoCl2 hypoxic conditions. Microscopy was used to assess cell morphology, while XTT, Trypan Blue, X-gal, LDH, Live Dead, scratch wound healing, and ROS assays were used to analyze viability, senescence, cell death, proliferation, and reactive oxygen species prevalence in the cells. According to our findings, CA priming enhances hASCs' ability to survive and regenerate in a hypoxic microenvironment more effectively than untreated hASCs. Our in-vitro research suggested that pre-treatment with CA of hASCs could be a unique way to enhance their therapeutic efficacy and ability to survive in hypoxic microenvironments.
Collapse
Affiliation(s)
- H.M. Shifa ul Haq
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | - Ramla Ashfaq
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
- Genome Editing Lab, Food Biotechnology Research Center, Pakistan Council of Scientific and Industrial Research (PCSIR) Laboratory Complex, Lahore, 54600, Pakistan
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | - Warda Shahid
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | - Ghufran Azam
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | - Maryam Azam
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | - Saba Tasneem
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | | | - Kausar Malik
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
| | - Sheikh Riazuddin
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of Punjab, Lahore, 53700, Pakistan
- Jinnah Burn & Reconstructive Surgery Centre, Allama Iqbal Medical College, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
14
|
Wan J, Wu T, Wang K, Xia K, Yin L, Chen C. Polydopamine-modified decellularized intestinal scaffolds loaded with adipose-derived stem cells promote intestinal regeneration. J Mater Chem B 2022; 11:154-168. [PMID: 36458582 DOI: 10.1039/d2tb01389d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Regeneration of gastrointestinal tissues remains a great challenge due to their unique microenvironment. Functional composite decellularized scaffolds have shown great potential in gastrointestinal repair and inducing gastrointestinal tissue-specific proliferation. In this study, polydopamine (PDA)-mediated surface modification of decellularized intestinal scaffolds (DIS), combined with adipose tissue-derived stem cells (ADSC), was used to promote intestinal wound healing while avoiding intestinal resection. The results showed that DIS had good biocompatibility and could maintain the growth and proliferation of ADSC. Moreover, PDA-coated DIS not only had anti-infection ability but could also further promote the secretory activity for the paracrine effects of ADSC. ADSC cultured on PDA-DIS produced significantly higher levels of anti-inflammatory and proangiogenic cytokines than those cultured on plastic plates or DIS. In vivo, ADSC-PDA-DIS significantly promoted intestinal wound closure in rat intestinal defect models. Moreover, ADSC-PDA-DIS was able to induce more neovascularization at 4 weeks postoperatively and promoted macrophage recruitment to accelerate wound healing. Taken together, the results showed that PDA-modified DIS could significantly improve the efficacy of stem cell therapy, and ADSC-PDA-DIS could improve the wound healing process with anti-infection effects, enhancing neovascularization and immunoregulation, which may be of great clinical significance for gastrointestinal regeneration.
Collapse
Affiliation(s)
- Jian Wan
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China. .,Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Tianqi Wu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Kai Xia
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Lu Yin
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
15
|
Vermeulen S, Van Puyvelde B, Bengtsson del Barrio L, Almey R, van der Veer BK, Deforce D, Dhaenens M, de Boer J. Micro-Topographies Induce Epigenetic Reprogramming and Quiescence in Human Mesenchymal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2203880. [PMID: 36414384 PMCID: PMC9811462 DOI: 10.1002/advs.202203880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Biomaterials can control cell and nuclear morphology. Since the shape of the nucleus influences chromatin architecture, gene expression and cell identity, surface topography can control cell phenotype. This study provides fundamental insights into how surface topography influences nuclear morphology, histone modifications, and expression of histone-associated proteins through advanced histone mass spectrometry and microarray analysis. The authors find that nuclear confinement is associated with a loss of histone acetylation and nucleoli abundance, while pathway analysis reveals a substantial reduction in gene expression associated with chromosome organization. In light of previous observations where the authors found a decrease in proliferation and metabolism induced by micro-topographies, they connect these findings with a quiescent phenotype in mesenchymal stem cells, as further shown by a reduction of ribosomal proteins and the maintenance of multipotency on micro-topographies after long-term culture conditions. Also, this influence of micro-topographies on nuclear morphology and proliferation is reversible, as shown by a return of proliferation when re-cultured on a flat surface. The findings provide novel insights into how biophysical signaling influences the epigenetic landscape and subsequent cellular phenotype.
Collapse
Affiliation(s)
- Steven Vermeulen
- Department of Instructive Biomaterials EngineeringMERLN InstituteUniversity of MaastrichtMaastricht6229 ERThe Netherlands
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Bart Van Puyvelde
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Laura Bengtsson del Barrio
- Department of Instructive Biomaterials EngineeringMERLN InstituteUniversity of MaastrichtMaastricht6229 ERThe Netherlands
| | - Ruben Almey
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Bernard K. van der Veer
- Laboratory for Stem Cell and Developmental EpigeneticsDepartment of Development and RegenerationKU LeuvenLeuven3000Belgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Maarten Dhaenens
- Laboratory of Pharmaceutical BiotechnologyDepartment of PharmaceuticsGhent UniversityGhent9000Belgium
| | - Jan de Boer
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
16
|
Peshkova M, Kosheleva N, Shpichka A, Radenska-Lopovok S, Telyshev D, Lychagin A, Li F, Timashev P, Liang XJ. Targeting Inflammation and Regeneration: Scaffolds, Extracellular Vesicles, and Nanotechnologies as Cell-Free Dual-Target Therapeutic Strategies. Int J Mol Sci 2022; 23:13796. [PMID: 36430272 PMCID: PMC9694395 DOI: 10.3390/ijms232213796] [Citation(s) in RCA: 9] [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: 09/16/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Osteoarthritis (OA) affects over 250 million people worldwide and despite various existing treatment strategies still has no cure. It is a multifactorial disease characterized by cartilage loss and low-grade synovial inflammation. Focusing on these two targets together could be the key to developing currently missing disease-modifying OA drugs (DMOADs). This review aims to discuss the latest cell-free techniques applied in cartilage tissue regeneration, since they can provide a more controllable approach to inflammation management than the cell-based ones. Scaffolds, extracellular vesicles, and nanocarriers can be used to suppress inflammation, but they can also act as immunomodulatory agents. This is consistent with the latest tissue engineering paradigm, postulating a moderate, controllable inflammatory reaction to be beneficial for tissue remodeling and successful regeneration.
Collapse
Affiliation(s)
- Maria Peshkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- FSBSI Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Anastasia Shpichka
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Stefka Radenska-Lopovok
- Institute for Clinical Morphology and Digital Pathology, Sechenov University, 119991 Moscow, Russia
| | - Dmitry Telyshev
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Moscow, Russia
- Institute of Bionic Technologies and Engineering, Sechenov University, 119991 Moscow, Russia
| | - Alexey Lychagin
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, 119991 Moscow, Russia
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Xing-Jie Liang
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Ti Y, Yang M, Chen X, Zhang M, Xia J, Lv X, Xiao D, Wang J, Lu M. Comparison of the therapeutic effects of human umbilical cord blood-derived mesenchymal stem cells and adipose-derived stem cells on erectile dysfunction in a rat model of bilateral cavernous nerve injury. Front Bioeng Biotechnol 2022; 10:1019063. [PMID: 36277409 PMCID: PMC9585154 DOI: 10.3389/fbioe.2022.1019063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Cavernous nerve injury (CNI) is the leading cause of erectile dysfunction (ED) after radical prostatectomy and pelvic fracture. Transplantation of human adipose-derived stem cells (ASCs) has been widely used to restore erectile function in CNI-ED rats and patients. Umbilical cord blood-derived MSCs (CBMSCs) are similarly low immunogenic but much primitive compared to ASCs and more promising in large-scale commercial applications due to the extensive establishment of cord blood banks. However, whether CBMSCs and ASCs have differential therapeutic efficacy on CNI-ED and the underlying mechanisms are still not clear. Materials and methods: A bilateral cavernous nerve injury (BCNI) rat model was established by crushing the bilateral cavernous nerves. After crushing, ASCs and CBMSCs were intracavernously injected immediately. Erectile function, Masson staining, and immunofluorescence analyses of penile tissues were assessed at 4 and 12 weeks. PKH-26-labeled ASCs or CBMSCs were intracavernously injected to determine the presence and differentiation of ASCs or CBMSCs in the penis 3 days after injection. In vitro experiments including intracellular ROS detection, mitochondrial membrane potential assay, EdU cell proliferation staining, cell apoptosis assay, and protein chip assay were conducted to explore the underlying mechanism of CBMSC treatment compared with ASC treatment. Results: CBMSC injection significantly restored erectile function, rescued the loss of cavernous corporal smooth muscles, and increased the ratio of smooth muscle to collagen. PKH-26-labeled CBMSCs or ASCs did not colocalize with endothelial cells or smooth muscle cells in the corpus cavernosum. Moreover, the conditioned medium (CM) of CBMSCs could significantly inhibit the oxidative stress and elevate the mitochondria membrane potential and proliferation of Schwann cells. Better therapeutic effects were observed in the CBMSC group than the ASC group both in vivo and in vitro. In addition, the content of neurotrophic factors and matrix metalloproteinases in CBMSC-CM, especially NT4, VEGF, MMP1, and MMP3 was significantly higher than that of ASC-CM. Conclusion: Intracavernous injection of CBMSCs exhibited a better erectile function restoration than that of ASCs in CNI-ED rats owing to richer secretory factors, which can promote nerve regeneration and reduce extracellular matrix deposition. CBMSC transplantation would be a promising therapeutic strategy for CNI-ED regeneration in the future.
Collapse
Affiliation(s)
- Yunrong Ti
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Mengbo Yang
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xinda Chen
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ming Zhang
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jingjing Xia
- Greater Bay Area Institute of Precision Medicine, School of Life Sciences, Fudan University, Guangzhou, China
| | - Xiangguo Lv
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dongdong Xiao
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Dongdong Xiao, ; Jiucun Wang, ; Mujun Lu,
| | - Jiucun Wang
- Greater Bay Area Institute of Precision Medicine, School of Life Sciences, Fudan University, Guangzhou, China
- Human Phenome Institute, Fudan University, Shanghai, China
- *Correspondence: Dongdong Xiao, ; Jiucun Wang, ; Mujun Lu,
| | - Mujun Lu
- Department of Urology and Andrology, Renji Hospital, Shanghai Institute of Andrology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Dongdong Xiao, ; Jiucun Wang, ; Mujun Lu,
| |
Collapse
|
18
|
Soares MBP, Gonçalves RGJ, Vasques JF, da Silva-Junior AJ, Gubert F, Santos GC, de Santana TA, Almeida Sampaio GL, Silva DN, Dominici M, Mendez-Otero R. Current Status of Mesenchymal Stem/Stromal Cells for Treatment of Neurological Diseases. Front Mol Neurosci 2022; 15:883378. [PMID: 35782379 PMCID: PMC9244712 DOI: 10.3389/fnmol.2022.883378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurological disorders include a wide spectrum of clinical conditions affecting the central and peripheral nervous systems. For these conditions, which affect hundreds of millions of people worldwide, generally limited or no treatments are available, and cell-based therapies have been intensively investigated in preclinical and clinical studies. Among the available cell types, mesenchymal stem/stromal cells (MSCs) have been widely studied but as yet no cell-based treatment exists for neurological disease. We review current knowledge of the therapeutic potential of MSC-based therapies for neurological diseases, as well as possible mechanisms of action that may be explored to hasten the development of new and effective treatments. We also discuss the challenges for culture conditions, quality control, and the development of potency tests, aiming to generate more efficient cell therapy products for neurological disorders.
Collapse
Affiliation(s)
- Milena B. P. Soares
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Renata G. J. Gonçalves
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J. da Silva-Junior
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Gubert
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Girlaine Café Santos
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Thaís Alves de Santana
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Gabriela Louise Almeida Sampaio
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | | | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Rosalia Mendez-Otero
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Shokravi S, Borisov V, Zaman BA, Niazvand F, Hazrati R, Khah MM, Thangavelu L, Marzban S, Sohrabi A, Zamani A. Mesenchymal stromal cells (MSCs) and their exosome in acute liver failure (ALF): a comprehensive review. Stem Cell Res Ther 2022; 13:192. [PMID: 35527304 PMCID: PMC9080215 DOI: 10.1186/s13287-022-02825-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 12/13/2022] Open
Abstract
Recently, mesenchymal stromal cells (MSCs) and their derivative exosome have become a promising approach in the context of liver diseases therapy, in particular, acute liver failure (ALF). In addition to their differentiation into hepatocytes in vivo, which is partially involved in liver regeneration, MSCs support liver regeneration as a result of their appreciated competencies, such as antiapoptotic, immunomodulatory, antifibrotic, and also antioxidant attributes. Further, MSCs-secreted molecules inspire hepatocyte proliferation in vivo, facilitating damaged tissue recovery in ALF. Given these properties, various MSCs-based approaches have evolved and resulted in encouraging outcomes in ALF animal models and also displayed safety and also modest efficacy in human studies, providing a new avenue for ALF therapy. Irrespective of MSCs-derived exosome, MSCs-based strategies in ALF include administration of native MSCs, genetically modified MSCs, pretreated MSCs, MSCs delivery using biomaterials, and also MSCs in combination with and other therapeutic molecules or modalities. Herein, we will deliver an overview regarding the therapeutic effects of the MSCs and their exosomes in ALF. As well, we will discuss recent progress in preclinical and clinical studies and current challenges in MSCs-based therapies in ALF, with a special focus on in vivo reports.
Collapse
Affiliation(s)
- Samin Shokravi
- Department of Research and Academic Affairs, Larkin Community Hospital, Miami, FL USA
| | - Vitaliy Borisov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Burhan Abdullah Zaman
- Basic Sciences Department, College of Pharmacy, University of Duhok, Duhok, Kurdistan Region Iraq
| | - Firoozeh Niazvand
- School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Raheleh Hazrati
- Department of Medicinal Chemistry, Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Meysam Mohammadi Khah
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Sima Marzban
- Department of Research and Academic Affairs, Larkin Community Hospital, Miami, FL USA
| | - Armin Sohrabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zamani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Teufelsbauer M, Lang C, Plangger A, Rath B, Moser D, Staud C, Radtke C, Neumayer C, Hamilton G. Effects of metformin on human bone-derived mesenchymal stromal cell-breast cancer cell line interactions. Med Oncol 2022; 39:54. [PMID: 35150338 PMCID: PMC8840908 DOI: 10.1007/s12032-022-01655-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
Metformin is used to treat patients with type 2 diabetes mellitus and was found to lower the incidence of cancer. Bone metastasis is a common impairment associated with advanced breast cancer. The present study investigated the effects of metformin on human bone-derived mesenchymal stromal cells (BM-MSC)—breast cancer cell line interactions. BM-MSCs grown from box chisels were tested for growth-stimulating and migration-controlling activity on four breast cancer cell lines either untreated or after pretreatment with metformin. Growth stimulation was tested in MTT tests and migration in scratch assays. Furthermore, the expression of adipokines of BM-MSCs in response to metformin was assessed using Western blot arrays. Compared to breast cancer cell lines (3.6 ± 1.4% reduction of proliferation), 500 µM metformin significantly inhibited the proliferation of BM-MSC lines (mean 12.3 ± 2.2 reduction). Pretreatment of BM-MSCs with metformin showed variable effects of the resulting conditioned media (CM) on breast cancer cell lines depending on the specific BM-MSC—cancer line combination. Metformin significantly reduced the migration of breast cancer cell lines MDA-MB-231 and MDA-MB-436 in response to CM of drug-pretreated BM-MSCs. Assessment of metformin-induced alterations in the expression of adipokines by BM-MSC CM indicated increased osteogenic signaling and possibly impairment of metastasis. In conclusion, the anticancer activities of metformin are the result of a range of direct and indirect mechanisms that lower tumor proliferation and progression. A lower metformin-induced protumor activity of BM-MSCs in the bone microenvironment seem to contribute to the positive effects of the drug in selected breast cancer patients.
Collapse
Affiliation(s)
- Maryana Teufelsbauer
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Clemens Lang
- Department of Trauma Surgery, Sozialmedizinisches Zentrum Ost, Donauspital, Vienna, Austria
| | - Adelina Plangger
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Doris Moser
- Department of Cranio, Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Neumayer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Lehmann J, Narcisi R, Franceschini N, Chatzivasileiou D, Boer CG, Koevoet WJLM, Putavet D, Drabek D, van Haperen R, de Keizer PLJ, van Osch GJVM, Ten Berge D. WNT/beta-catenin signalling interrupts a senescence-induction cascade in human mesenchymal stem cells that restricts their expansion. Cell Mol Life Sci 2022; 79:82. [PMID: 35048158 PMCID: PMC8770385 DOI: 10.1007/s00018-021-04035-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/18/2021] [Accepted: 11/09/2021] [Indexed: 12/23/2022]
Abstract
Senescence, the irreversible cell cycle arrest of damaged cells, is accompanied by a deleterious pro-inflammatory senescence-associated secretory phenotype (SASP). Senescence and the SASP are major factors in aging, cancer, and degenerative diseases, and interfere with the expansion of adult cells in vitro, yet little is known about how to counteract their induction and deleterious effects. Paracrine signals are increasingly recognized as important senescence triggers and understanding their regulation and mode of action may provide novel opportunities to reduce senescence-induced inflammation and improve cell-based therapies. Here, we show that the signalling protein WNT3A counteracts the induction of paracrine senescence in cultured human adult mesenchymal stem cells (MSCs). We find that entry into senescence in a small subpopulation of MSCs triggers a secretome that causes a feed-forward signalling cascade that with increasing speed induces healthy cells into senescence. WNT signals interrupt this cascade by repressing cytokines that mediate this induction of senescence. Inhibition of those mediators by interference with NF-κB or interleukin 6 signalling reduced paracrine senescence in absence of WNT3A and promoted the expansion of MSCs. Our work reveals how WNT signals can antagonize senescence and has relevance not only for expansion of adult cells but can also provide new insights into senescence-associated inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Johannes Lehmann
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Center for Molecular Medicine, Section Molecular Cancer Research, Division LAB, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Natasja Franceschini
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Danai Chatzivasileiou
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Cindy G Boer
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Wendy J L M Koevoet
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Diana Putavet
- Center for Molecular Medicine, Section Molecular Cancer Research, Division LAB, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Dubravka Drabek
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Harbour Biomed, Rotterdam, the Netherlands
| | - Rien van Haperen
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Harbour Biomed, Rotterdam, the Netherlands
| | - Peter L J de Keizer
- Center for Molecular Medicine, Section Molecular Cancer Research, Division LAB, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Gerjo J V M van Osch
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Derk Ten Berge
- Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Guo X, Wang J, Zou W, Wei W, Guan X, Liu J. Exploring microenvironment strategies to delay mesenchymal stem cell senescence. Stem Cells Dev 2021; 31:38-52. [PMID: 34913751 DOI: 10.1089/scd.2021.0254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as an important candidate for cell therapy and tissue regeneration. However, some limitations in translational research and therapies still exist, such as insufficient cell supply, inadequate differentiation potential, and decreased immune capacity, all of which result from replicative senescence during long-term in vitro culture. In vitro, stem cells lack a protective microenvironment owing to the absence of physical and biochemical cues compared with the in vivo niche, which provides dynamic physicochemical and biological cues. This difference results in accelerated aging after long-term in vitro culture. Therefore, it remains a great challenge to delay replicative senescence in culture. Constructing a microenvironment to delay replicative senescence of MSCs by maintaining their phenotypes, properties, and functions is a feasible strategy to solve this problem and has made measurable progress both in preclinical studies and clinical trials. Here, we review the current knowledge on the characteristics of senescent MSCs, explore the molecular mechanisms of MSCs senescence, describe the niche of MSCs, and discuss some current microenvironment strategies to delay MSCs replicative senescence that can broaden their range of therapeutic applications.
Collapse
Affiliation(s)
- Xunhui Guo
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, China;
| | - Jiayi Wang
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, Dalian, China;
| | - Wei Zou
- Liaoning Normal University, 66523, College of Life Sciences, Dalian, China;
| | - Wenjuan Wei
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Xin Guan
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Jing Liu
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| |
Collapse
|
23
|
Huang P, Zhang C, Delawary M, Korchak JA, Suda K, Zubair AC. Development and evaluation of IL-6 overexpressing mesenchymal stem cells (MSCs). J Tissue Eng Regen Med 2021; 16:244-253. [PMID: 34890489 DOI: 10.1002/term.3274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/08/2022]
Abstract
Mesenchymal stem/stromal cell (MSC) therapy has been investigated in multiple diseases and conditions. Although the mechanisms of MSC-based therapies are not fully understood, we and others have shown interleukin 6 (IL-6) to be an important factor in MSC function. IL-6 contributes to many biological events, such as immune response, neurogenesis, and bone remodeling. In our study, we tested the feasibility of engineering MSCs by IL-6 mRNA transfection (eMSCs-IL6) and evaluated the optimal time to harvest them after transfection. We then assessed the functional characteristics of eMSCs-IL6. Quantitative real-time PCR and ELISA results have shown that mature IL-6 mRNA was efficiently transfected into MSCs using a lipofectamine based method. The IL-6 mRNA and protein overexpression peaked after 1 day of transfection and the secreted IL-6 protein was sustained for at least 6 days. A short time course experiment demonstrated that 4 h after transfection was the best time point to harvest and freeze eMSCs-IL6 for future studies. In addition, eMSCs-IL6 maintained their characteristics as defined by International Society for Cell & Gene Therapy. The immunosuppressive capacity of conditioned culture medium (CCM) from eMSCs-IL6 (CCM-IL6) was significantly enhanced compared to naïve MSCs conditioned culture medium (CCM-control). Our studies established for the first time the feasibility of efficiently generating IL-6 overexpressing MSCs which have enhanced immunosuppressive capacity. This is providing a novel approach to improve the efficacy of MSCs for potential application in regenerative medicine.
Collapse
Affiliation(s)
- Peng Huang
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Cuiping Zhang
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Mina Delawary
- Cell Therapy Research Laboratories, Daiichi Sankyo, Co, Ltd, Tokyo, Japan
| | - Jennifer A Korchak
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Koji Suda
- Cell Therapy Research Laboratories, Daiichi Sankyo, Co, Ltd, Tokyo, Japan
| | - Abba C Zubair
- Center for Regenerative Medicine and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
24
|
Mirhaj M, Tavakoli M, Varshosaz J, Labbaf S, Jafarpour F, Ahmaditabar P, Salehi S, Kazemi N. Platelet rich fibrin containing nanofibrous dressing for wound healing application: Fabrication, characterization and biological evaluations. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112541. [DOI: 10.1016/j.msec.2021.112541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/25/2021] [Accepted: 11/06/2021] [Indexed: 12/27/2022]
|
25
|
Kim S, Jung PY, Lee JS, Hwang S, Sohn JH, Yoon Y, Bae KS, Eom YW. Cultured human skeletal muscle satellite cells exhibit characteristics of mesenchymal stem cells and play anti-inflammatory roles through prostaglandin E2 and hepatocyte growth factors. Cell Biol Int 2021; 45:2443-2451. [PMID: 34374483 DOI: 10.1002/cbin.11688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/06/2021] [Accepted: 08/07/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle satellite cells (SkMSCs) play crucial roles in muscle fiber maintenance, repair, and remodeling; however, it remains unknown if these properties are preserved in cultured SkMSCs. In this study, we investigated the characteristics of cultured SkMSCs and their ability to regulate the activity of M1 macrophages. SkMSCs grew well with an average population doubling time of 26.26 ± 6.85 h during 10 passages (P). At P5, Pax7, MyoD, cluster of differentiation (CD)34, and CD56 were not expressed in SkMSCs, but the MSC markers CD73, CD105, and CD90 were expressed and the cells were differentiated into adipocytes and osteoblasts. When SkMSCs were cocultured with macrophages, interleukin (IL)-1β secretion was decreased, prostaglandin (PG)E2 was produced in coculture, and cyclooxygenase-2 protein was induced in an SkMSC-dependent manner. Hepatocyte growth factor (HGF) was highly secreted by monocultured SkMSCs; interferon-γ and lipopolysaccharide reduced its expression level. However, HGF expression recovered when SkMSCs and macrophages were cocultured. Although exogenous PGE2 upregulated macrophage pro-IL-1β expression, it suppressed the secretion of cleaved IL-1β. In contrast, HGF decreased active IL-1β secretion without affecting pro-IL-1β expression. Co-treatment of macrophages with HGF and PGE2 reduced pro-IL-1β expression level and active IL-1β secretion. Our results suggest that SkMSCs lose their satellite cell properties during serial passaging but acquire mesenchymal stem cell properties including the ability to exert an anti-inflammatory response for macrophages through PGE2 and HGF.
Collapse
Affiliation(s)
- Seongyup Kim
- Department of General Surgery, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Pil Young Jung
- Department of General Surgery, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Jin Suk Lee
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Soonjae Hwang
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Joon Hyung Sohn
- Central Research Laboratory, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Yongdae Yoon
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Keum Seok Bae
- Department of General Surgery, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Young Woo Eom
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.,Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| |
Collapse
|
26
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
27
|
Rastegar A, Mahmoodi M, Mirjalili M, Nasirizadeh N. Platelet-rich fibrin-loaded PCL/chitosan core-shell fibers scaffold for enhanced osteogenic differentiation of mesenchymal stem cells. Carbohydr Polym 2021; 269:118351. [PMID: 34294355 DOI: 10.1016/j.carbpol.2021.118351] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/27/2021] [Accepted: 06/13/2021] [Indexed: 01/18/2023]
Abstract
Here, we fabricated the platelet-rich fibrin (PRF)-loaded PCL/chitosan (PCL/CS-PRF) core-shell nanofibrous scaffold through a coaxial electrospinning method. Our goal was to evaluate the effect of CS-RPF in the core layer of the nanofibrous on the osteogenic differentiation of human mesenchymal stem cells (HMSCs). The elastic modulus of PCL/CS-PRF core-shell scaffold (44 MPa) was about 1.5-fold of PCL/CS scaffold (25 MPa). The specific surface area of the scaffolds increased from 9.98 m2/g for PCL/CS scaffold to 16.66 m2/g for the PCL/CS-PRF core-shell nanofibrous scaffold. Moreover, the release rate of PRF from PCL/CS-PRF nanofibrous scaffold was measured to be 24.50% after 10 days which showed slow and sustained release of PRF from the nanofibrous. The formation of Ca-P on the surface of scaffold immersed in simulated body fluid solution indicated the suitable osteoconductivity of PCL/CS-PRF core-shell nanofibrous scaffold. Also, the value of ALP activity and calcium deposited on the surface of PCL/CS-PRF core-shell nanofibrous scaffold were 81.97 U/L and 40.33 μg/scaffold, respectively after 14 days, which confirmed the significantly higher amounts of ALP and calcium deposition on the scaffold containing PRF compared to PCL/CS scaffold. Due to higher hydrophilicity and porosity of PCL/CS-PRF core-shell nanofibrous scaffold compared to PCL/CS scaffold, a better bone cell growth on surface of PCL/CS-PRF scaffold was observed. The Alizarin red-positive area was significantly higher on PCL/CS-PRF scaffold compared to PCL/CS scaffold, indicating more calcium deposition and osteogenic differentiation of HMSCs in the presence of PRF. Our findings demonstrate that PCL/CS-PRF core-shell scaffolds can provide a strong construct with improved osteogenic for bone tissue engineering applications.
Collapse
Affiliation(s)
- Amirabbas Rastegar
- Department of Textile Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Mahboobeh Mahmoodi
- Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran.
| | - Mohammad Mirjalili
- Department of Textile Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Navid Nasirizadeh
- Department of Chemical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| |
Collapse
|
28
|
Towards Physiologic Culture Approaches to Improve Standard Cultivation of Mesenchymal Stem Cells. Cells 2021; 10:cells10040886. [PMID: 33924517 PMCID: PMC8069108 DOI: 10.3390/cells10040886] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest for their use in cell-based therapies due to their multipotent differentiation and immunomodulatory capacities. In consequence of limited numbers following their isolation from the donor tissue, MSCs require extensive expansion performed in traditional 2D cell culture setups to reach adequate amounts for therapeutic use. However, prolonged culture of MSCs in vitro has been shown to decrease their differentiation potential and alter their immunomodulatory properties. For that reason, preservation of these physiological characteristics of MSCs throughout their in vitro culture is essential for improving the efficiency of therapeutic and in vitro modeling applications. With this objective in mind, many studies already investigated certain parameters for enhancing current standard MSC culture protocols with regard to the effects of specific culture media components or culture conditions. Although there is a lot of diversity in the final therapeutic uses of the cells, the primary stage of standard isolation and expansion is imperative. Therefore, we want to review on approaches for optimizing standard MSC culture protocols during this essential primary step of in vitro expansion. The reviewed studies investigate and suggest improvements focused on culture media components (amino acids, ascorbic acid, glucose level, growth factors, lipids, platelet lysate, trace elements, serum, and xenogeneic components) as well as culture conditions and processes (hypoxia, cell seeding, and dissociation during passaging), in order to preserve the MSC phenotype and functionality during the primary phase of in vitro culture.
Collapse
|
29
|
Yoshitomi T, Zheng H, Yoshimoto K. Investigations of Chirality Effects on Undifferentiated State of Mesenchymal Stem Cells Using Soft Nanofibrous Oligopeptide Hydrogels. ANAL SCI 2021; 37:539-543. [PMID: 33281137 DOI: 10.2116/analsci.20scn05] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Soft nanofibrous oligopeptide hydrogels consisting of self-assembled l- and d-form Fmoc-Phe-Phe-Cys networks photo-cross-linked by poly(ethylene glycol)-dimethacrylate, which are referred to as l- and d-gel, respectively, were developed for investigation of chirality effects on the undifferentiated state of mesenchymal stem cells. Encapsulated mesenchymal stem cells in d-gel showed slower growth and less spreading, resulting in higher maintenance of the undifferentiated state, compared to in l-gel. This indicates that d-form peptide materials might be useful as scaffold materials for regenerative medicine using stem cells.
Collapse
Affiliation(s)
- Toru Yoshitomi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo.,Research Center for Functional Materials, National Institute for Materials Science
| | - Hangyu Zheng
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo.,Present Address: Institute for Frontier Life and Medical Sciences, Kyoto University
| | - Keitaro Yoshimoto
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo
| |
Collapse
|
30
|
Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochem 2020; 122:151627. [PMID: 33002788 DOI: 10.1016/j.acthis.2020.151627] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/17/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
Spermatogonial stem cells (SSCs) are very sensitive to chemotherapy and radiotherapy, so male infertility is a great challenge for prepubertal cancer survivors. Cryoconservation of testicular cells before cancer treatment can preserve SSCs from treatment side effects. Different two-dimensional (2D) and three-dimensional (3D) culture systems of SSCs have been used in many species as a useful technique to in vitro spermatogenesis. We evaluated the proliferation of SSCs in 2D and 3D culture systems of platelet-rich plasma (PRP). testicular cells of four brain-dead patients cultivated in 2D pre-culture system, characterization of SSCs performed by RT-PCR, flow cytometry, immunocytochemistry and their functionality assessed by xenotransplantation to azoospermia mice. PRP prepared and dosimetry carried out to determine the optimized dose of PRP. After preparation of PRP scaffold, cytotoxic and histological evaluation performed and SSCs cultivated into three groups: control, 2D culture by optimized dose of PRP and PRP scaffold. The diameter and number of colonies measured and relative expression of GFRa1 and c-KIT evaluated by real-time PCR. Results indicated the expression of PLZF, VASA, OCT4, GFRa1 and vimentin in colonies after 2D pre-culture, xenotransplantation demonstrated proliferated SSCs have proper functionality to homing in mouse testes. The relative expression of c-KIT showed a significant increase as compared to the control group (*: p < 0.05) in PRP- 2D group, expression of GFRa1 and c-KIT in PRP scaffold group revealed a significant increase as compared to other groups (***: p < 0.001). The number and diameter of colonies in the PRP-2D group showed a considerable increase (p < 0.01) as compared to the control group. In PRP- scaffold group, a significant increase (p < 0.01) was seen only in the number of colonies related to the control group. Our results suggested that PRP scaffold can reconstruct a suitable structure to the in vitro proliferation of SSCs.
Collapse
|
31
|
Lim YL, Eom YW, Park SJ, Hong T, Kang SH, Baik SK, Park KS, Kim MY. Bone Marrow-Derived Mesenchymal Stem Cells Isolated from Patients with Cirrhosis and Healthy Volunteers Show Comparable Characteristics. Int J Stem Cells 2020; 13:394-403. [PMID: 32840228 PMCID: PMC7691862 DOI: 10.15283/ijsc20072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
Background and Objectives Autologous or allogeneic bone marrow-derived mesenchymal stem cells (BMSCs) have been applied in clinical trials to treat liver disease. However, only a few studies are comparing the characteristics of autologous MSCs from patients and allogeneic MSCs from normal subjects. Methods and Results We compared the characteristics of BMSCs (BCs and BPs, respectively) isolated from six healthy volunteers and six patients with cirrhosis. In passage 3 (P3), senescent population and expression of p53 and p21 were slightly higher in BPs, but the average population doubling time for P3–P5 in BPs was approximately 65.3±11.1 h, which is 18.4 h shorter than that in BCs (83.7±9.2 h). No difference was observed in the expression of CD73, CD90, or CD105 between BCs and BPs. Adipogenic differentiation slightly increased in BCs, but the expression levels of leptin, peroxisome proliferator-activated receptor γ, and CCAAT-enhancer-binding protein α did not vary between differentiated BCs and BPs. While ATP and reactive oxygen species levels were slightly lower in BPs, mitochondrial membrane potential, oxygen consumption rate, and expression of mitochondria-related genes such as cytochrome c oxidase 1 were not significantly different between BCs and BPs. Conclusions Taken together, there are marginal differences in the proliferation, differentiation, and mitochondrial activities of BCs and BPs, but both BMSCs from patients with cirrhosis and healthy volunteers show comparable characteristics.
Collapse
Affiliation(s)
- Yoo Li Lim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Su Jung Park
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Taeui Hong
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seong Hee Kang
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Soon Koo Baik
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kyu-Sang Park
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
32
|
Toulmonde M, Lucchesi C, Verbeke S, Crombe A, Adam J, Geneste D, Chaire V, Laroche-Clary A, Perret R, Bertucci F, Bertolo F, Bianchini L, Dadone-Montaudie B, Hembrough T, Sweet S, Kim YJ, Cecchi F, Le Loarer F, Italiano A. High throughput profiling of undifferentiated pleomorphic sarcomas identifies two main subgroups with distinct immune profile, clinical outcome and sensitivity to targeted therapies. EBioMedicine 2020; 62:103131. [PMID: 33254023 PMCID: PMC7708794 DOI: 10.1016/j.ebiom.2020.103131] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 10/01/2020] [Accepted: 10/29/2020] [Indexed: 01/15/2023] Open
Abstract
Background Undifferentiated pleomorphic sarcoma (UPS) is the most frequent, aggressive and less-characterized sarcoma subtype. This study aims to assess UPS molecular characteristics and identify specific therapeutic targets. Methods High-throughput technologies encompassing immunohistochemistry, RNA-sequencing, whole exome-sequencing, mass spectrometry, as well as radiomics were used to characterize three independent cohorts of 110, 25 and 41 UPS selected after histological review performed by an expert pathologist. Correlations were made with clinical outcome. Cell lines and xenografts were derived from human samples for functional experiments. Findings CD8 positive cell density was independently associated with metastatic behavior and prognosis. RNA-sequencing identified two main groups: the group A, enriched in genes involved in development and stemness, including FGFR2, and the group B, strongly enriched in genes involved in immunity. Immune infiltrate patterns on tumor samples were highly predictive of gene expression classification, leading to call the group B ‘immune-high’ and the group A ‘immune-low’. This molecular classification and its prognostic impact were confirmed on an independent cohort of UPS from TCGA. Copy numbers alterations were significantly more frequent in immune-low UPS. Proteomic analysis identified two main proteomic groups that highly correlated with the two main transcriptomic groups. A set of nine radiomic features from conventional MRI sequences provided the basis for a radiomics signature that could select immune-high UPS on their pre-therapeutic imaging. Finally, in vitro and in vivo anti-tumor activity of FGFR inhibitor JNJ-42756493 was selectively shown in cell lines and patient-derived xenograft models derived from immune-low UPS. Interpretation Two main disease entities of UPS, with distinct immune phenotypes, prognosis, molecular features and MRI textures, as well as differential sensitivity to specific anticancer agents were identified. Immune-high UPS may be the best candidates for immune checkpoint inhibitors, whereas this study provides rational for assessing FGFR inhibition in immune-low UPS. Funding This work was partly founded by a grant from La Ligue.
Collapse
Affiliation(s)
- Maud Toulmonde
- Medical Oncology Department, Institut Bergonié, Bordeaux, France; University of Bordeaux, Bordeaux, France
| | - Carlo Lucchesi
- Bioinformatics Department, Institut Bergonié, Bordeaux, France; INSERM U1218, Bordeaux, France
| | - Stéphanie Verbeke
- INSERM U1218, Bordeaux, France; Research Department, Institut Bergonié, Bordeaux, France
| | - Amandine Crombe
- University of Bordeaux, Bordeaux, France; Radiology Department, Institut Bergonié, Bordeaux, France
| | - Julien Adam
- Pathology Department, Gustave Roussy, Villejuif, France
| | - Damien Geneste
- Bioinformatics Department, Institut Bergonié, Bordeaux, France; INSERM U1218, Bordeaux, France
| | - Vanessa Chaire
- INSERM U1218, Bordeaux, France; Research Department, Institut Bergonié, Bordeaux, France
| | - Audrey Laroche-Clary
- Bioinformatics Department, Institut Bergonié, Bordeaux, France; Research Department, Institut Bergonié, Bordeaux, France
| | - Raul Perret
- Pathology Department, Institut Bergonié, Bordeaux, France
| | - François Bertucci
- Centre de Recherche en Cancérologie de Marseille (CRCM), CNRS UMR725, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Frederic Bertolo
- Bioinformatics Department, Institut Bergonié, Bordeaux, France; INSERM U1218, Bordeaux, France
| | - Laurence Bianchini
- Laboratory of solid tumor genetics, Université Côte d'Azur (UCA), CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Bérengère Dadone-Montaudie
- Laboratory of solid tumor genetics, Université Côte d'Azur (UCA), CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | | | | | | | | | | | - Antoine Italiano
- Medical Oncology Department, Institut Bergonié, Bordeaux, France; University of Bordeaux, Bordeaux, France; INSERM U1218, Bordeaux, France.
| |
Collapse
|
33
|
Sid-Otmane C, Perrault LP, Ly HQ. Mesenchymal stem cell mediates cardiac repair through autocrine, paracrine and endocrine axes. J Transl Med 2020; 18:336. [PMID: 32873307 PMCID: PMC7466793 DOI: 10.1186/s12967-020-02504-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
In the past decade, despite key advances in therapeutic strategies following myocardial infarction, none can directly address the loss of cardiomyocytes following ischemic injury. Cardiac cell-based therapy is at the cornerstone of regenerative medicine that has shown potential for tissue repair. Mesenchymal stem cells (MSC) represent a strong candidate to heal the infarcted myocardium. While differentiation potential has been described as a possible avenue for MSC-based repair, their secreted mediators are responsible for the majority of the ascribed prohealing effects. MSC can either promote their own survival and proliferation through autocrine effect or secrete trophic factors that will act on adjacent cells through a paracrine effect. Prior studies have also documented beneficial effects even when MSCs were remotely delivered, much akin to an endocrine mechanism. This review aims to distinguish the paracrine activity of MSCs from an endocrine-like effect, where remotely transplanted cells can promote healing of the injured myocardium.
Collapse
Affiliation(s)
- Celia Sid-Otmane
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.,Research Centre, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada
| | - Louis P Perrault
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.,Research Centre, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Cardiovascular Surgery, Montreal Heart Institute and Université de Montréal, Montreal, QC, Canada
| | - Hung Q Ly
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada. .,Department of Medicine, Université de Montréal, Montreal, QC, Canada. .,Research Centre, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.
| |
Collapse
|
34
|
Johnstone BH, Miller HM, Beck MR, Gu D, Thirumala S, LaFontaine M, Brandacher G, Woods EJ. Identification and characterization of a large source of primary mesenchymal stem cells tightly adhered to bone surfaces of human vertebral body marrow cavities. Cytotherapy 2020; 22:617-628. [PMID: 32873509 PMCID: PMC8919862 DOI: 10.1016/j.jcyt.2020.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/12/2020] [Accepted: 07/05/2020] [Indexed: 12/13/2022]
Abstract
Background: Therapeutic allogeneic mesenchymal stromal cells (MSCs) are currently in clinical trials to evaluate their effectiveness in treating many different disease indications. Eventual commercialization for broad distribution will require further improvements in manufacturing processes to economically manufacture MSCs at scales sufficient to satisfy projected demands. A key contributor to the present high cost of goods sold for MSC manufacturing is the need to create master cell banks from multiple donors, which leads to variability in large-scale manufacturing runs. Therefore, the availability of large single donor depots of primary MSCs would greatly benefit the cell therapy market by reducing costs associated with manufacturing. Methods: We have discovered that an abundant population of cells possessing all the hallmarks of MSCs is tightly associated with the vertebral body (VB) bone matrix and only liberated by proteolytic digestion. Here we demonstrate that these vertebral bone-adherent (vBA) MSCs possess all the International Society of Cell and Gene Therapy-defined characteristics (e.g., plastic adherence, surface marker expression and trilineage differentiation) of MSCs, and we have therefore termed them vBA-MSCs to distinguish this population from loosely associated MSCs recovered through aspiration or rinsing of the bone marrow compartment. Results: Pilot banking and expansion were performed with vBA-MSCs obtained from 3 deceased donors, and it was demonstrated that bank sizes averaging 2.9 × 108 ± 1.35 × 108 vBA-MSCs at passage 1 were obtainable from only 5 g of digested VB bone fragments. Each bank of cells demonstrated robust proliferation through a total of 9 passages, without significant reduction in population doubling times. The theoretical total cell yield from the entire amount of bone fragments (approximately 300 g) from each donor with limited expansion through 4 passages is 100 trillion (1 × 1014) vBA-MSCs, equating to over 105 doses at 10 × 106 cells/kg for an average 70-kg recipient. Discussion: Thus, we have established a novel and plentiful source of MSCs that will benefit the cell therapy market by overcoming manufacturing and regulatory inefficiencies due to donor-to-donor variability.
Collapse
Affiliation(s)
- Brian H Johnstone
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA.
| | - Hannah M Miller
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA
| | - Madelyn R Beck
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dongsheng Gu
- Ossium Health, Inc, Indianapolis, Indiana, USA; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sreedhar Thirumala
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael LaFontaine
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erik J Woods
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
35
|
Mesenchymal Stem/Progenitor Cells: The Prospect of Human Clinical Translation. Stem Cells Int 2020; 2020:8837654. [PMID: 33953753 PMCID: PMC8063852 DOI: 10.1155/2020/8837654] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/19/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) are key players in regenerative medicine, relying principally on their differentiation/regeneration potential, immunomodulatory properties, paracrine effects, and potent homing ability with minimal if any ethical concerns. Even though multiple preclinical and clinical studies have demonstrated remarkable properties for MSCs, the clinical applicability of MSC-based therapies is still questionable. Several challenges exist that critically hinder a successful clinical translation of MSC-based therapies, including but not limited to heterogeneity of their populations, variability in their quality and quantity, donor-related factors, discrepancies in protocols for isolation, in vitro expansion and premodification, and variability in methods of cell delivery, dosing, and cell homing. Alterations of MSC viability, proliferation, properties, and/or function are also affected by various drugs and chemicals. Moreover, significant safety concerns exist due to possible teratogenic/neoplastic potential and transmission of infectious diseases. Through the current review, we aim to highlight the major challenges facing MSCs' human clinical translation and shed light on the undergoing strategies to overcome them.
Collapse
|
36
|
Li M, Jiang T, Zhang W, Xie W, Guo T, Tang X, Zhang J. Human umbilical cord MSC-derived hepatocyte growth factor enhances autophagy in AOPP-treated HK-2 cells. Exp Ther Med 2020; 20:2765-2773. [PMID: 32765771 PMCID: PMC7401891 DOI: 10.3892/etm.2020.8998] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation may serve as an important treatment modality in chronic kidney disease (CKD); however, the underlying mechanisms remain unclear. Advanced oxidation protein products (AOPP) have been demonstrated to induce renal tubular epithelial cell (RTEC) injury via autophagy inhibition. Therefore, the present study was performed to investigate the role of human umbilical cord-derived MSCs (hUC-MSCs) in RTEC autophagy. AOPP-treated HK-2 cells were co-cultured with hUC-MSCs or treated with recombinant humanized hepatocyte growth factor (HGF). Western blotting was used to detect the levels of autophagy-and PI3K/AKT/mTOR signaling pathway-related proteins, and immunofluorescence staining was used to detect the levels of autophagy-related proteins. The HGF protein levels in HK-2 cells and the hUC-MSC co-culture system were measured. The cells were subsequently treated with tivantinib, an HGF competitive inhibitor, and the levels of autophagy-related proteins were detected. Microtubule-associated protein 1 light chain 3B (LC3B) II/LC3B I (LC3II/LC3I) and beclin 1 protein levels were increased, while p62, PI3K, phosphorylated (p)-AKT and the p-mTOR protein levels were decreased in AOPP-treated HK-2 cells co-cultured with hUC-MSC, compared with the group treated with AOPP only. Furthermore, HGF expression was increased in AOPP-treated HK-2 cells co-cultured with hUC-MSC, compared with the group treated with AOPP alone. When HGF activity was inhibited using tivantinib, these effects on LC3II/LC3I, beclin 1, p62, PI3K, p-AKT, and p-mTOR expression were partially reversed. Furthermore, the effects of tivantinib were reversed by Ly294002. In conclusion, the present study revealed that hUC-MSCs partially reversed AOPP-mediated inhibition of autophagy in HK-2 cells via secretion of HGF, indicating that hUC-MSCs may serve as a potential therapy for preventing the progression of CKD.
Collapse
Affiliation(s)
- Minhui Li
- Blood Purification Center, Nanhai Hospital of Southern Medical University, Foshan, Guangdong 528244, P.R. China
| | - Tingting Jiang
- Department of Nephrology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Wenying Zhang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Wei Xie
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Tingting Guo
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Xun Tang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Jun Zhang
- Department of Nephrology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
37
|
Fujimoto Y, Yokozeki T, Yokoyama A, Tabata Y. Basic fibroblast growth factor enhances proliferation and hepatocyte growth factor expression of feline mesenchymal stem cells. Regen Ther 2020; 15:10-17. [PMID: 32490062 PMCID: PMC7256438 DOI: 10.1016/j.reth.2020.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/26/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction The objective of this study is to evaluate the effect of basic fibroblast growth factor (bFGF) on the proliferation and secretion activity of feline adipose-derived mesenchymal stem cells (MSC). Methods Feline MSC isolated from the subcutaneous adipose tissue of cats were cultured with or without bFGF. Results The bFGF addition enhanced the proliferation of feline MSC to a significant great extent compared with that without bFGF, although the cell proliferation tended to increase with the bFGF concentration. In addition, adipogenic and osteogenic staining assay demonstrated that the bFGF addition allowed MSC to maintain the differentiation ability even after the proliferation. Moreover, no change in the surface markers of MSC was observed between the cultures with or without bFGF. A quantitative RT-PCR assay revealed that the HGF and TSG-6 expression significantly increased by the bFGF addition. The highest mRNA expression of MMP-2 was observed for cells cultured in 1000 ng/ml bFGF concentration. Conclusions The culture with bFGF is a promising way to enhance the proliferation, and HGF secretion ability of MSC as well as maintain their differentiation ability and immunophenotype nature. Feline adipose-derived mesenchymal stem cells (MSC) was cultured with or without the basic fibroblast growth factor (bFGF). The bFGF enhanced the proliferation and increased the mRNA expression of HGF, TSG-6, and MMP-2. The bFGF addition was not influenced to the differentiation ability and cell surface marker of MSC.
Collapse
Key Words
- Basic fibroblast growth factor
- CKD, chronic kidney disease
- ECM, extracellular matrix
- FBS, fetal bovine serum
- FGF, basic fibroblast growth factor
- Feline
- GAPDH, gliyceraldehyde-3-phosphate dehydrogenase
- HGF, hepatocyte growth factor
- Hepatocyte growth factor
- MMP-2, matrix metalloproteinase-2
- MSC, mesenchymal stem cells
- Mesenchymal stem cell
- P1, passage 1
- Proliferation
- SVF, stromal vascular fraction
- TSG-6, tumor necrosis factor-stimulated gene 6
- Tumor necrosis factor-stimulated gene 6
Collapse
Affiliation(s)
- Youhei Fujimoto
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan.,Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
38
|
Mehrian M, Lambrechts T, Papantoniou I, Geris L. Computational Modeling of Human Mesenchymal Stromal Cell Proliferation and Extra-Cellular Matrix Production in 3D Porous Scaffolds in a Perfusion Bioreactor: The Effect of Growth Factors. Front Bioeng Biotechnol 2020; 8:376. [PMID: 32411692 PMCID: PMC7201129 DOI: 10.3389/fbioe.2020.00376] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 04/06/2020] [Indexed: 01/20/2023] Open
Abstract
Stem cell expansion on 3D porous scaffolds cultured in bioreactor systems has been shown to be beneficial for maintenance of the original cell functionality in tissue engineering strategies (TE). However, the production of extracellular matrix (ECM) makes harvesting the progenitor cell population from 3D scaffolds a challenge. Medium composition plays a role in stimulating cell proliferation over extracellular matrix (ECM) production. In this regard, a computational model describing tissue growth inside 3D scaffolds can be a great tool in designing optimal experimental conditions. In this study, a computational model describing cell and ECM growth in a perfusion bioreactor is developed, including a description of the effect of a (generic) growth factor on the biological processes taking place inside the 3D scaffold. In the model, the speed of cell and ECM growth depends on the flow-induced shear stress, curvature and the concentrations of oxygen, glucose, lactate, and growth factor. The effect of the simulated growth factor is to differentially enhance cell proliferation over ECM production. After model calibration with historic in-house data, a multi-objective optimization procedure is executed aiming to minimize the total experimental cost whilst maximizing cell growth during culture. The obtained results indicate there are multiple optimum points for the medium refreshment regime and the initial growth factor concentration where a trade-off is made between the final amount of cells and the culture cost. Finally, the model is applied to experiments reported in the literature studying the effects of perfusion-based cell culture and/or growth factor supplementation on cell expansion. The qualitative similarities between the simulation and experimental results, even in the absence of proper model calibration, reinforces the generic character of the proposed modeling framework. The model proposed in this study can contribute to the cost efficient production of cell-based TE products, ultimately contributing to their affordability and accessibility.
Collapse
Affiliation(s)
- Mohammad Mehrian
- Biomechanics Research Unit, GIGA In silico Medicine, University of Liège, Liège, Belgium.,Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Toon Lambrechts
- Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,M3-BIORES, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Institute of Chemical Engineering Sciences (ICEHT), Foundation for Research and Technology - Hellas (FORTH), Patras, Greece
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In silico Medicine, University of Liège, Liège, Belgium.,Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Liu G, Wu R, Yang B, Shi Y, Deng C, Atala A, Mou S, Criswell T, Zhang Y. A cocktail of growth factors released from a heparin hyaluronic-acid hydrogel promotes the myogenic potential of human urine-derived stem cells in vivo. Acta Biomater 2020; 107:50-64. [PMID: 32044457 DOI: 10.1016/j.actbio.2020.02.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/22/2020] [Accepted: 02/04/2020] [Indexed: 01/19/2023]
Abstract
Traditional cell therapy technology relies on the maximum expansion of primary stem cells in vitro, through multiple passages and potential differentiation protocols, in order to generate the abundance of cells needed prior to transplantation in vivo. Implantation of in vitro over-expanded and pre-differentiated cells typically results in poor cell survival and reduced regeneration capacity for tissue repair in vivo. We hypothesized that implantation of primary stem cells, after a short time culture in vitro (passage number ≤p3), in combination with controlled release of relevant growth factors would improve in vivo cell viability, engraftment and tissue regeneration. The goal of this study was to determine whether the release of myogenic growth factors from a heparin-hyaluronic acid gel (hp-HA gel) could enhance in vivo cell survival, in-growth and myogenic differentiation of human urine-derived stem cells (USC) with a corresponding enhancement in graft vascularization, innervation and regenerative properties. Human USC were obtained from healthy adult donors (n = 6), expanded and then mixed with a hp-HA gel containing sets of growth factors known to enhance myogenesis (IGF1, HGF, PDGF-BB), neurogenesis (NGF, FGF) and angiogenesis (VEGF), or a cocktail with a combination of growth factors. Primary cultured USC (p3) mixed with the hp-HA gel and the various combinations of growth factors, were subcutaneously injected into athymic mice. In vivo cell survival, engraftment and functional differentiation within the host tissue were assessed. The implanted grafts containing USC and the growth factor cocktail showed the greatest number of surviving cells as well as increased numbers of cells that expressed myogenic and endothelial cell markers as compared to other groups 4 weeks after implantation. Moreover, the graft with USC and the growth factor cocktail showed increased numbers of blood vessels and infiltrating neurons. Thus, growth factors released in a controlled manner from an hp-HA gel containing USC efficiently improved in vivo cell survival and supported vascularization and myogenic differentiation within the grafts. This study provides evidence for the use of primary USC and growth factors in a hydrogel as a novel mode of cell therapy for the promotion of myogenic differentiation for the treatment of injured muscle tissue. STATEMENT OF SIGNIFICANCE: Cell therapies are a promising treatment option for neuromuscular dysfunction disorders. However, major limitations in cell retention and engraftment after implantation remain a hindrance to the use of stem cell therapy for the treatment of muscle injuries or diseased tissues. Implanted long-term in vitro cultured cells tend to demonstrate low rates of survival and tissue engraftment, lessened paracrine effects, and poor homing and differentiation. Human USC are an easily obtainable stem cell source that possess stem cell characteristics such as a robust proliferative potential, paracrine effects on neighboring cells, and multi-potential differentiation. In this study, we demonstrated that a combination of primary human USC with a cocktail of growth factors combined in a hyaluronic gel was optimal for cell survival and engraftment, including myogenic differentiation potential of USC, angiogenesis and host nerve fiber recruitment in vivo. The present study also demonstrated that the use of primary urine derived stem cells at early passages, without in vitro pre-differentiation, implanted in a hyaluronic-heparin hydrogel containing a cocktail of growth factors, provided an alternative safe site-specific delivery method for cell therapy.
Collapse
Affiliation(s)
- Guihua Liu
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rongpei Wu
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bin Yang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yingai Shi
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Chunhua Deng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Steven Mou
- Anesthesiology-Pediatric ICU Anesthesia at WakeForest Baptist Medical Center, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Tracy Criswell
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
40
|
Cheng Y, Lin K, Young T, Cheng N. The influence of fibroblast growth factor 2 on the senescence of human adipose-derived mesenchymal stem cells during long-term culture. Stem Cells Transl Med 2020; 9:518-530. [PMID: 31840944 PMCID: PMC7103622 DOI: 10.1002/sctm.19-0234] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) exhibit great potential in regenerative medicine, and in vitro expansion is frequently necessary to obtain a sufficient number of ASCs for clinical use. Fibroblast growth factor 2 (FGF2) is a common supplement in the ASC culture medium to enhance cell proliferation. To achieve clinical applicability of ASC-based products, prolonged culture of ASCs is sometimes required to obtain sufficient quantity of ASCs. However, the effect of FGF2 on ASCs during prolonged culture has not been previously determined. In this study, ASCs were subjected to prolonged in vitro culture with or without FGF2. FGF2 maintained the small cell morphology and expedited proliferation kinetics in early ASC passages. After prolonged in vitro expansion, FGF2-treated ASCs exhibited increased cell size, arrested cell proliferation, and increased cellular senescence relative to the control ASCs. We observed an upregulation of FGFR1c and enhanced expression of downstream STAT3 in the initial passages of FGF2-treated ASCs. The application of an FGFR1 or STAT3 inhibitor effectively blocked the enhanced proliferation of ASCs induced by FGF2 treatment. FGFR1c upregulation and enhanced STAT3 expression were lost in the later passages of FGF2-treated ASCs, suggesting that the continuous stimulation of FGF2 becomes ineffective because of the refractory downstream FGFR1 and the STAT3 signaling pathway. In addition, no evidence of tumorigenicity was noted in vitro and in vivo after prolonged expansion of FGF2-cultured ASCs. Our data indicate that ASCs have evolved a STAT3-dependent response to continuous FGF2 stimulation which promotes the initial expansion but limits their long-term proliferation.
Collapse
Affiliation(s)
- Yin Cheng
- Department of SurgeryNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Kai‐Hsuan Lin
- Department of SurgeryNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Tai‐Horng Young
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - Nai‐Chen Cheng
- Department of SurgeryNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
- Research Center for Developmental Biology and Regenerative MedicineNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
41
|
Li J, Song S, Li X, Zhu J, Li W, Du B, Guo Y, Xi X, Han R. Down-Regulation of Fibroblast Growth Factor 2 (FGF2) Contributes to the Premature Senescence of Mouse Embryonic Fibroblast. Med Sci Monit 2020; 26:e920520. [PMID: 32188838 PMCID: PMC7104787 DOI: 10.12659/msm.920520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Freshly isolated mouse embryonic fibroblasts (MEFs) have great proliferation capacity but quickly enter senescent state after several rounds of cell cycle, a process called premature senescence. Cellular senescence can be induced by various stresses such as telomere erosion, DNA damage, and oncogenic signaling. But the contribution of other molecules, such as growth factors, to cellular senescence is incompletely understood. This study aimed to compare the gene expression difference between non-senescent and senescent MEFs to identify the key molecule(s) involved in the spontaneous senescence of MEFs. Material/Methods Primary MEFs were isolated from E12.5 pregnant C57/BL6 mice. The cells were continuously cultured in Dulbecco’s Modified Eagle Medium for 9 passages. SA-β-Gal staining was used as an indicator of cell senescence. The supernatant from primary MEFs (P1 medium) or Passage 6 MEFs (P6 medium) were used to culture freshly isolated MEFs to observe the effects on cell senescence state. Gene expression profiles of primary and senescent MEFs were investigated by RNA-Seq to find the key genes involved in cell senescence. Adipocyte differentiation assay was used to evaluate the stemness of MEFs cultured in FGF2-stimulated medium. Results The senescence of MEFs cultured in the P1 medium was alleviated when compared to the P6 medium. Downregulation of FGF2 expression was revealed by RNA-Seq and further confirmed by real-time quantitative polymerase chain reaction and western blot. FGF2-stimulated medium also had anti-senescence function and could maintain the differentiation ability of MEFs. Conclusions The premature senescence of MEFs was at least partially caused by FGF2 deficiency. Exogenous FGF2 could alleviate the senescent phenotype.
Collapse
Affiliation(s)
- Jie Li
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Shuo Song
- Department of Science and Education, Shenzhen Samii Medical Center, Shenzhen, Guangdong, China (mainland)
| | - Xingchao Li
- Department of Pediatric Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Jing Zhu
- Department of Biology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China (mainland).,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Wenjuan Li
- Department of Pharmacology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Boyu Du
- Department of Biology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Yang Guo
- Department of Immunology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Xueyan Xi
- Department of Immunology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Rongfei Han
- Department of Immunology, School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China (mainland).,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China (mainland).,Department of Laboratory Medicine, Suizhou Central Hospital, Suizhou, Hubei, China (mainland)
| |
Collapse
|
42
|
Mohanty C, Pradhan J. A human epidermal growth factor-curcumin bandage bioconjugate loaded with mesenchymal stem cell for in vivo diabetic wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110751. [PMID: 32279771 DOI: 10.1016/j.msec.2020.110751] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/16/2020] [Accepted: 02/15/2020] [Indexed: 12/24/2022]
Abstract
Bone-marrow-derived mesenchymal stem cells (MSCs) are of growing interest for the treatment of diabetic wound healing. However, they are often associated with poor proliferation and viability at the wounded site. Here, it is reported the use of human epidermal growth factor -curcumin bandage bioconjugate (EGF-Cur B) loaded with MSCs (MSCs-EGF-Cur B) at the wounded site for diabetic wound healing. Conjugation efficiency of EGF was determined by FTIR and XPS, surface morphology was analyzed by SEM and AFM and hydrophilicity by contact angle. Chemical integrity of curcumin with the polymeric matrix was studied by FTIR and, antiinflamatory and biocompatibility of EGF-Cur B were determined by TNF α ELISA and MTT study respectively. The culture of MSCs over EGF-Cur B enhanced MSC viability and expression of transcription factors associated with the maintenance of pluripotency and self-renewal (OCT¾, SOX2, and Nanog) as compared to MSCs grown in standard conditions. Its therapeutic effect was examined on diabetic full-thickness excisional wound model in terms of size and histological examination. Synergetic combinational approach especially when treated with MSCs-EGF-Cur B significantly enhanced wound closure by increasing granulation tissue formation, collagen deposition, and angiogenesis as compared to other groups. In conclusion, biocompatible therapeutic MSCs-EGF-Cur B might have great application for diabetic wound healing in the near future.
Collapse
Affiliation(s)
- Chandana Mohanty
- Institute of Life Sciences, Nalco Square, Bhubaneswar, India; School of Applied Science, KIIT University, Bhubaneswar, Odisha, India.
| | | |
Collapse
|
43
|
The fate of mesenchymal stem cells is greatly influenced by the surface chemistry of silica nanoparticles in 3D hydrogel-based culture systems. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 106:110259. [DOI: 10.1016/j.msec.2019.110259] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 12/17/2022]
|
44
|
Park SR, Kim SR, Park CH, Lim S, Ha SY, Hong IS, Lee HY. Sonic Hedgehog, a Novel Endogenous Damage Signal, Activates Multiple Beneficial Functions of Human Endometrial Stem Cells. Mol Ther 2019; 28:452-465. [PMID: 31866117 DOI: 10.1016/j.ymthe.2019.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 01/20/2023] Open
Abstract
Local endometrial stem cells play an important role in regulating endometrial thickness, which is an essential factor for successful embryo implantation and pregnancy outcomes. Importantly, defects in endometrial stem cell function can be responsible for thin endometrium and subsequent recurrent pregnancy losses. Therefore, many researchers have directed their efforts toward finding a novel stimulatory factor that can enhance the regenerative capacity of endometrial stem cells. Sonic hedgehog (SHH) is a morphogen that plays a key role in regulating pattern formation throughout embryonic limb development. In addition to this canonical function, we identified for the first time that SHH is actively secreted as a stem cell-activating factor in response to tissue injury and subsequently stimulates tissue regeneration by promoting various beneficial functions of endometrial stem cells. Our results also showed that SHH exerts stimulatory effects on endometrial stem cells via the FAK/ERK1/2 and/or phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways. More importantly, we also observed that endometrial stem cells stimulated with SHH showed markedly enhanced differentiation and migratory capacities and subsequent in vivo therapeutic effects in an endometrial ablation animal model.
Collapse
Affiliation(s)
- Se-Ra Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Soo-Rim Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Chan Hum Park
- Department of Otolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, South Korea
| | - Soyi Lim
- Department of Obstetrics and Gynecology, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Seung Yeon Ha
- Department of Pathology, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea; Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea.
| | - Hwa-Yong Lee
- Department of Biomedical Science, Jungwon University, 85 Goesan-eup, Munmu-ro, Goesan-gun, Chungcheongbuk-do 367-700, Republic of Korea.
| |
Collapse
|
45
|
Therapeutic effect of mesenchymal stem cells derived from human umbilical cord in rabbit temporomandibular joint model of osteoarthritis. Sci Rep 2019; 9:13854. [PMID: 31554894 PMCID: PMC6761110 DOI: 10.1038/s41598-019-50435-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/12/2019] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative condition of the temporomandibular joint (TMJ) characterised by chronic inflammation and damage to joint structures. Because of the complexity of TMJ-OA, only symptomatic treatments are currently available. Recent reports have shown that many of stem cells can exert anti-inflammatory and tissue-regenerating effects. In this study, we investigated the potential cartilage-regenerating and anti-inflammatory effects of human umbilical cord matrix-mesenchymal stem cells (hUCM-MSCs) for the treatment of TMJ-OA. hUCM-MSC lines, isolated from different donors, which showed different activities in vitro. Using a selected cell line, we used different concentrations of hUCM-MSCs to assess therapeutic effects in a rabbit model of monosodium iodoacetate-induced TMJ-OA. Compared with the untreated control group, the potential regenerative result and anti-inflammatory effects of hUCM-MSCs were evident at all the tested concentrations in rabbits with induced TMJ-OA. The median dose of hUCM-MSCs showed the prominent cartilage protective effect and further cartilage regeneration potential. This effect occurred via upregulated expression of growth factors, extracellular matrix markers, and anti-inflammatory cytokines, and reduced expression of pro-inflammatory cytokines. The anti-inflammatory effect of hUCM-MSCs was comparable to that of dexamethasone (DEX). However, only hUCM-MSCs showed potential chondrogenesis effects in this study. In conclusion, our results indicate that hUCM-MSCs may be an effective treatment option for the treatment of TMJ-OA.
Collapse
|
46
|
Jung JS, Volk C, Marga C, Navarrete Santos A, Jung M, Rujescu D, Navarrete Santos A. Adipose-Derived Stem/Stromal Cells Recapitulate Aging Biomarkers and Show Reduced Stem Cell Plasticity Affecting Their Adipogenic Differentiation Capacity. Cell Reprogram 2019; 21:187-199. [PMID: 31298565 DOI: 10.1089/cell.2019.0010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stromal mesenchymal stem cells (MSCs) have the capability to self-renew and can differentiate into multiple cell types of the mesoderm germ layer, but their properties are affected by molecular aging mechanisms. MSCs can be obtained from adipose tissue termed as adipose-derived stem/stromal cells (ASCs) representing a promising tool for studying age-related diseases in detail. ASCs from young (16 weeks) and old (>108 weeks) rabbits were successfully isolated and propagated. ASCs showed the typical morphology and stained positive for CD105, Vimentin, Collagenase 1A, and negative for CD14, CD90, and CD73, demonstrating their mesenchymal origin. ASCs expressed MSC markers, including MYC, KLF4, CHD1, REST, and KAT6A, whereas pluripotency-related genes, such as NANOG, OCT4, and SOX2, were not expressed. Aged ASCs showed altered protein and mRNA levels of APOE, ATG7, FGF2, PTEN, and SIRT1. Adipogenic differentiation of old visceral ASCs was significantly decreased compared with young visceral ASCs. We successfully established rabbit ASC cultures representing an in vitro model for the analysis of stem cell aging mechanisms. ASCs, obtained from old female rabbits, showed age- and source-specific alteration due to aging of the donor. Stem cell plasticity was altered with age as shown by reduced adipogenic differentiation capacity.
Collapse
Affiliation(s)
- Juliane-Susanne Jung
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Christin Volk
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Christina Marga
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Alexander Navarrete Santos
- 2Center for Medical Basic Research, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Matthias Jung
- 3Department of Psychiatry, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Dan Rujescu
- 3Department of Psychiatry, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Anne Navarrete Santos
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| |
Collapse
|
47
|
Mattinzoli D, Ikehata M, Tsugawa K, Alfieri CM, Barilani M, Lazzari L, Andreetta P, Elli FM, Mantovani G, Messa P. FGF23 and Fetuin-A Interaction and Mesenchymal Osteogenic Transformation. Int J Mol Sci 2019; 20:ijms20040915. [PMID: 30791553 PMCID: PMC6412477 DOI: 10.3390/ijms20040915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/08/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022] Open
Abstract
Recently, we found a strict bone association between Fibroblast growth factor 23 (FGF23) and Fetuin-A, both involved in cardiovascular and mineral bone disorders. In this study, an uninvestigated bone marrow positivity for both was found. Though the role of exogenous FGF23 on mesenchymal cells (MSCs) was reported, no information is as yet available on the possible production of this hormone by MSCs. To further analyze these uninvestigated aspects, we studied human primary cells and mouse and human cell lines by means of immunostaining, qRT-PCR, enzyme linked immunosorbent assays, chromatin immunoprecipitation, transfection, and a streamlined approach for the FGF23⁻Fetuin-A interaction called Duolink proximity ligation assay. Mesenchymal cells produce but do not secrete FGF23 and its expression increases during osteo-differentiation. Fibroblast growth factor 23 is also involved in the regulation of Fetuin-A by binding directly to the Fetuin-A promoter and then activating its transcription. Both FGF23 overexpression and addition induced an upregulation of Fetuin-A in the absence of osteo-inducer factors. Fibroblast growth factor 23 and Fetuin-A promoter were increased by osteo-inducer factors with this effect being abolished after FGF23 silencing. In conclusion, both FGF23 and Fetuin-A are present and strictly linked to each other in MSCs with FGF23 driving Fetuin-A production. This mechanism suggests a role for these two proteins in the osteoblast differentiation.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Masami Ikehata
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Koji Tsugawa
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Carlo M Alfieri
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
- Unit of Nephrology, Dialysis and Renal transplant Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Mario Barilani
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy.
- Department of Transfusion Medicine and Hematology, Cell Factory, Regenerative medicine laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Lorenza Lazzari
- Department of Transfusion Medicine and Hematology, Cell Factory, Regenerative medicine laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Paola Andreetta
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Francesca M Elli
- Department of Clinical Sciences and Community Health, Endocrinology Unit, University of Milan, 20122 Milan, Italy.
- Unit of Endocrinology and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, Endocrinology Unit, University of Milan, 20122 Milan, Italy.
- Unit of Endocrinology and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Piergiorgio Messa
- Renal Research Laboratory Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
- Unit of Nephrology, Dialysis and Renal transplant Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy.
| |
Collapse
|
48
|
Hamann A, Nguyen A, Pannier AK. Nucleic acid delivery to mesenchymal stem cells: a review of nonviral methods and applications. J Biol Eng 2019; 13:7. [PMID: 30675180 PMCID: PMC6339289 DOI: 10.1186/s13036-019-0140-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are multipotent stem cells that can be isolated and expanded from many tissues, and are being investigated for use in cell therapies. Though MSC therapies have demonstrated some success, none have been FDA approved for clinical use. MSCs lose stemness ex vivo, decreasing therapeutic potential, and face additional barriers in vivo, decreasing therapeutic efficacy. Culture optimization and genetic modification of MSCs can overcome these barriers. Viral transduction is efficient, but limited by safety concerns related to mutagenicity of integrating viral vectors and potential immunogenicity of viral antigens. Nonviral delivery methods are safer, though limited by inefficiency and toxicity, and are flexible and scalable, making them attractive for engineering MSC therapies. Main text Transfection method and nucleic acid determine efficiency and expression profile in transfection of MSCs. Transfection methods include microinjection, electroporation, and nanocarrier delivery. Microinjection and electroporation are efficient, but are limited by throughput and toxicity. In contrast, a variety of nanocarriers have been demonstrated to transfer nucleic acids into cells, however nanocarrier delivery to MSCs has traditionally been inefficient. To improve efficiency, plasmid sequences can be optimized by choice of promoter, inclusion of DNA targeting sequences, and removal of bacterial elements. Instead of DNA, RNA can be delivered for rapid protein expression or regulation of endogenous gene expression. Beyond choice of nanocarrier and nucleic acid, transfection can be optimized by priming cells with media additives and cell culture surface modifications to modulate barriers of transfection. Media additives known to enhance MSC transfection include glucocorticoids and histone deacetylase inhibitors. Culture surface properties known to modulate MSC transfection include substrate stiffness and specific protein coating. If nonviral gene delivery to MSCs can be sufficiently improved, MSC therapies could be enhanced by transfection for guided differentiation and reprogramming, transplantation survival and directed homing, and secretion of therapeutics. We discuss utilized delivery methods and nucleic acids, and resulting efficiency and outcomes, in transfection of MSCs reported for such applications. Conclusion Recent developments in transfection methods, including nanocarrier and nucleic acid technologies, combined with chemical and physical priming of MSCs, may sufficiently improve transfection efficiency, enabling scalable genetic engineering of MSCs, potentially bringing effective MSC therapies to patients.
Collapse
Affiliation(s)
- Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE 68583-0726 USA
| | - Albert Nguyen
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE 68583-0726 USA
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE 68583-0726 USA
| |
Collapse
|
49
|
Hsu YK, Sheu SY, Wang CY, Chuang MH, Chung PC, Luo YS, Huang JJ, Ohashi F, Akiyoshi H, Kuo TF. The effect of adipose-derived mesenchymal stem cells and chondrocytes with platelet-rich fibrin releasates augmentation by intra-articular injection on acute osteochondral defects in a rabbit model. Knee 2018; 25:1181-1191. [PMID: 30420268 DOI: 10.1016/j.knee.2018.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/25/2018] [Accepted: 10/10/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND This study aimed to investigate the efficacy of adipose-derived mesenchymal stem cells (ADSCs), platelet-rich fibrin releasates (PRFr), and chondrocyte transplantation in rabbit acute osteochondral defects. METHODS Thirty rabbits were randomly assigned to five groups: untreated controls; ADSCs alone; PRFr alone; PRFr + ADSCs; and PRFr + chondrocytes. The critical size osteochondral defects in right knee femoral condyles were injected intra-articularly according to the groups, as listed. The experimental rabbits received treatments once a week for two weeks postoperatively. All evaluations were conducted for 14 weeks following surgery, and the regenerated cartilages were assessed by gross inspection and histological examination. RESULTS There were no complications encountered in any of the rabbits. The size of the defect decreased and the volume of repaired cartilage increased in the medial femoral condyles of the PRFr + ADSCs group. Relative to the ADSCs or PRFr group, histological examination demonstrated that the PRFr + ADSCs group had thicker hyaline cartilage-specific extracellular matrix. Grading scores revealed that PRFr + ADSCs injection had better matrix, cell distribution, and surface indices than other groups (P < 0.05). However, the histological scores reported for PRFr + chondrocytes on cartilage repair were similar to those of PRFr, and there were no significant between-group differences. CONCLUSIONS These findings showed that intra-articular injections of PRFr + ADSCs into the knee can reduce cartilage defects by regenerating hyaline-like cartilage without complications. This approach may provide an alternative method for functional reconstruction of acute osteochondral defects with an unlimited source of cells and releasates.
Collapse
Affiliation(s)
- Yuan-Kai Hsu
- Veterinary Surgery, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Shi-Yuan Sheu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Integrated Chinese and Western Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Hsi Chuang
- Ph.D. Program of Technology Management, Chung Hwa University, Hsinchu, Taiwan; Gwo Xi Stem Cell Applied Technology Co., Ltd., Hsinchu, Taiwan
| | - Pei-Chun Chung
- School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Siang Luo
- School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Jun-Jie Huang
- School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fumihito Ohashi
- Veterinary Surgery, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Hideo Akiyoshi
- Veterinary Surgery, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Tzong-Fu Kuo
- School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; Department of Post-Baccalaureate Veterinary Medicine, Asia University, Taichung, Taiwan.
| |
Collapse
|
50
|
Hassanpour M, Rezabakhsh A, Pezeshkian M, Rahbarghazi R, Nouri M. Distinct role of autophagy on angiogenesis: highlights on the effect of autophagy in endothelial lineage and progenitor cells. Stem Cell Res Ther 2018; 9:305. [PMID: 30409213 PMCID: PMC6225658 DOI: 10.1186/s13287-018-1060-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autophagy plays a critical role in the dynamic growth of each cell through different conditions. It seems that this intracellular mechanism acts as a two-edged sword against the numerous cell insults. Previously, autophagy was described in the context of cell activity and behavior, but little knowledge exists related to the role of autophagy in endothelial cells, progenitors, and stem cells biology from different tissues. Angiogenic behavior of endothelial lineage and various stem cells are touted as an inevitable feature in the restoration of different damaged tissues and organs. This capacity was found to be dictated by autophagy signaling pathway. This review article highlights the fundamental role of cell autophagic response in endothelial cells function, stem cells dynamic, and differentiation rate. It seems that elucidation of the mechanisms related to pro- and/or anti-angiogenic potential of autophagy inside endothelial cells and stem cells could help us to modulate stem cell therapeutic feature post-transplantation.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756 Iran
- Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Emergency Medicine Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Pezeshkian
- Department of Applied Drug Research, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756 Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756 Iran
| |
Collapse
|