1
|
Zhou Y, Cai X, Zhang X, Dong Y, Pan X, Lai M, Zhang Y, Chen Y, Li X, Li X, Liu J, Zhang Y, Ma F. Mesenchymal stem/stromal cells from human pluripotent stem cell-derived brain organoid enhance the ex vivo expansion and maintenance of hematopoietic stem/progenitor cells. Stem Cell Res Ther 2024; 15:68. [PMID: 38443990 PMCID: PMC10916050 DOI: 10.1186/s13287-023-03624-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/22/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Mesenchymal stem/stromal cells (MSCs) are of great therapeutic value due to their role in maintaining the function of hematopoietic stem/progenitor cells (HSPCs). MSCs derived from human pluripotent stem cells represent an ideal alternative because of their unlimited supply. However, the role of MSCs with neural crest origin derived from HPSCs on the maintenance of HSPCs has not been reported. METHODS Flow cytometric analysis, RNA sequencing and differentiation ability were applied to detect the characteristics of stromal cells from 3D human brain organoids. Human umbilical cord blood CD34+ (UCB-CD34+) cells were cultured in different coculture conditions composed of stromal cells and umbilical cord MSCs (UC-MSCs) with or without a cytokine cocktail. The hematopoietic stroma capacity of stromal cells was tested in vitro with the LTC-IC assay and in vivo by cotransplantation of cord blood nucleated cells and stroma cells into immunodeficient mice. RNA and proteomic sequencing were used to detect the role of MSCs on HSPCs. RESULTS The stromal cells, derived from both H1-hESCs and human induced pluripotent stem cells forebrain organoids, were capable of differentiating into the classical mesenchymal-derived cells (osteoblasts, chondrocytes, and adipocytes). These cells expressed MSC markers, thus named pluripotent stem cell-derived MSCs (pMSCs). The pMSCs showed neural crest origin with CD271 expression in the early stage. When human UCB-CD34+ HSPCs were cocultured on UC-MSCs or pMSCs, the latter resulted in robust expansion of UCB-CD34+ HSPCs in long-term culture and efficient maintenance of their transplantability. Comparison by RNA sequencing indicated that coculture of human UCB-CD34+ HSPCs with pMSCs provided an improved microenvironment for HSC maintenance. The pMSCs highly expressed the Wnt signaling inhibitors SFRP1 and SFRP2, indicating that they may help to modulate the cell cycle to promote the maintenance of UCB-CD34+ HSPCs by antagonizing Wnt activation. CONCLUSIONS A novel method for harvesting MSCs with neural crest origin from 3D human brain organoids under serum-free culture conditions was reported. We demonstrate that the pMSCs support human UCB-HSPC expansion in vitro in a long-term culture and the maintenance of their transplantable ability. RNA and proteomic sequencing indicated that pMSCs provided an improved microenvironment for HSC maintenance via mechanisms involving cell-cell contact and secreted factors and suppression of Wnt signaling. This represents a novel method for large-scale production of MSCs of neural crest origin and provides a potential approach for development of human hematopoietic stromal cell therapy for treatment of dyshematopoiesis.
Collapse
Affiliation(s)
- Ya Zhou
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Xinping Cai
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College(CAMS & PUMC), Tianjin, 300020, China
| | - Xiuxiu Zhang
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Yong Dong
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Xu Pan
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Mowen Lai
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Yimeng Zhang
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Yijin Chen
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Xiaohong Li
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Xia Li
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Jiaxin Liu
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China
| | - Yonggang Zhang
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China.
| | - Feng Ma
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Huacai Road 26, Chengdu, 610052, China.
| |
Collapse
|
2
|
iPSC-Derived MSCs Are a Distinct Entity of MSCs with Higher Therapeutic Potential than Their Donor-Matched Parental MSCs. Int J Mol Sci 2023; 24:ijms24010881. [PMID: 36614321 PMCID: PMC9821152 DOI: 10.3390/ijms24010881] [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: 12/07/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stromal cells derived from induced pluripotent stem cells (iMSCs) have been proposed as alternative sources of primary MSCs with various advantages for cell therapeutic trials. However, precise evaluation of the differences between iMSCs and primary MSCs is lacking due to individual variations in the donor cells, which obscure direct comparisons between the two. In this study, we generated donor-matched iMSCs from individual bone marrow-derived MSCs and directly compared their cell-autonomous and paracrine therapeutic effects. We found that the transition from primary MSCs to iMSCs is accompanied by a functional shift towards higher proliferative activity, with variations in differentiation potential in a donor cell-dependent manner. The transition from MSCs to iMSCs was associated with common changes in transcriptomic and proteomic profiles beyond the variations of their individual donors, revealing expression patterns unique for the iMSCs. These iMSC-specific patterns were characterized by a shift in cell fate towards a pericyte-like state and enhanced secretion of paracrine cytokine/growth factors. Accordingly, iMSCs exhibited higher support for the self-renewing expansion of primitive hematopoietic progenitors and more potent immune suppression of allogenic immune responses than MSCs. Our study suggests that iMSCs represent a separate entity of MSCs with unique therapeutic potential distinct from their parental MSCs, but points to the need for iMSC characterization in the individual basis.
Collapse
|
3
|
Oliveira CS, Carreira M, Correia CR, Mano JF. The Therapeutic Potential of Hematopoietic Stem Cells in Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:379-392. [PMID: 33683146 DOI: 10.1089/ten.teb.2021.0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The repair process of bone fractures is a complex biological mechanism requiring the recruitment and in situ functionality of stem/stromal cells from the bone marrow (BM). BM mesenchymal stem/stromal cells have been widely explored in multiple bone tissue engineering applications, whereas the use of hematopoietic stem cells (HSCs) has been poorly investigated in this context. A reasonable explanation is the fact that the role of HSCs and their combined effect with other elements of the hematopoietic niches in the bone-healing process is still elusive. Therefore, in this review we intend to highlight the influence of HSCs in the bone repair process, mainly through the promotion of osteogenesis and angiogenesis at the bone injury site. For that, we briefly describe the main biological characteristics of HSCs, as well as their hematopoietic niches, while reviewing the biomimetic engineered BM niche models. Moreover, we also highlighted the role of HSCs in translational in vivo transplantation or implantation as promoters of bone tissue repair.
Collapse
Affiliation(s)
- Cláudia S Oliveira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Mariana Carreira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Clara R Correia
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
4
|
Sfougataki I, Varela I, Stefanaki K, Karagiannidou A, Roubelakis MG, Kalodimou V, Papathanasiou I, Traeger-Synodinos J, Kitsiou-Tzeli S, Kanavakis E, Kitra V, Tsezou A, Tzetis M, Goussetis E. Proliferative and chondrogenic potential of mesenchymal stromal cells from pluripotent and bone marrow cells. Histol Histopathol 2020; 35:1415-1426. [PMID: 32959885 DOI: 10.14670/hh-18-259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) can be derived from a wide range of fetal and adult sources including pluripotent stem cells (PSCs). The properties of PSC-derived MSCs need to be fully characterized, in order to evaluate the feasibility of their use in clinical applications. PSC-MSC proliferation and differentiation potential in comparison with bone marrow (BM)-MSCs is still under investigation. The objective of this study was to determine the proliferative and chondrogenic capabilities of both human induced pluripotent stem cell (hiPSC-) and embryonic stem cell (hESC-) derived MSCs, by comparing them with BM-MSCs. METHODS MSCs were derived from two hiPSC lines (hiPSC-MSCs), the well characterized Hues9 hESC line (hESC-MSCs) and BM from two healthy donors (BM-MSCs). Proliferation potential was investigated using appropriate culture conditions, with serial passaging, until cells entered into senescence. Differentiation potential to cartilage was examined after in vitro chondrogenic culture conditions. RESULTS BM-MSCs revealed a fold expansion of 1.18x10⁵ and 2.3x10⁵ while the two hiPSC-MSC lines and hESC-MSC showed 5.88x10¹⁰, 3.49x10⁸ and 2.88x10⁸, respectively. Under chondrogenic conditions, all MSC lines showed a degree of chondrogenesis. However, when we examined the formed chondrocyte micromasses by histological analysis of the cartilage morphology and immunohistochemistry for the chondrocyte specific markers Sox9 and Collagen II, we observed that PSC-derived MSC lines had formed pink rather than hyaline cartilage, in contrast to BM-MSCs. CONCLUSION In conclusion, MSCs derived from both hESCs and hiPSCs had superior proliferative capacity compared to BM-MSCs, but they were inefficient in their ability to form hyaline cartilage.
Collapse
Affiliation(s)
- Irene Sfougataki
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece.,Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Aghia Sophia Children's Hospital, Athens, Greece.
| | - Ioanna Varela
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece
| | - Kalliope Stefanaki
- Department of Histopathology, Aghia Sophia Children's Hospital, Athens, Greece
| | | | - Maria G Roubelakis
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Kalodimou
- Flow Cytometry-Research and Regenerative Medicine Department, IASO Hospital, Athens, Greece
| | - Ioanna Papathanasiou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessally, Thessally, Greece
| | - Joanne Traeger-Synodinos
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Kitsiou-Tzeli
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanuel Kanavakis
- Genesis Genoma Lab, Genetic diagnosis, Clinical Genetics and Research, Chalandri, Greece
| | - Vasiliki Kitra
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessally, Thessally, Greece
| | - Maria Tzetis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evgenios Goussetis
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece
| |
Collapse
|
5
|
Lee Chong T, Ahearn EL, Cimmino L. Reprogramming the Epigenome With Vitamin C. Front Cell Dev Biol 2019; 7:128. [PMID: 31380368 PMCID: PMC6646595 DOI: 10.3389/fcell.2019.00128] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/02/2019] [Indexed: 12/19/2022] Open
Abstract
The erasure of epigenetic modifications across the genome of somatic cells is an essential requirement during their reprogramming into induced pluripotent stem cells (iPSCs). Vitamin C plays a pivotal role in remodeling the epigenome by enhancing the activity of Jumonji-C domain-containing histone demethylases (JHDMs) and the ten-eleven translocation (TET) proteins. By maintaining differentiation plasticity in culture, vitamin C also improves the quality of tissue specific stem cells derived from iPSCs that are highly sought after for use in regenerative medicine. The ability of vitamin C to potentiate the activity of histone and DNA demethylating enzymes also has clinical application in the treatment of cancer. Vitamin C deficiency has been widely reported in cancer patients and has recently been shown to accelerate cancer progression in disease models. Therapies involving high-dose vitamin C administration are currently gaining traction in the treatment of epigenetic dysregulation, by targeting aberrant histone and DNA methylation patterns associated with cancer progression.
Collapse
Affiliation(s)
- Taylor Lee Chong
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Emily L Ahearn
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Luisa Cimmino
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
6
|
Abdal Dayem A, Lee SB, Kim K, Lim KM, Jeon TI, Seok J, Cho ASG. Production of Mesenchymal Stem Cells Through Stem Cell Reprogramming. Int J Mol Sci 2019; 20:ijms20081922. [PMID: 31003536 PMCID: PMC6514654 DOI: 10.3390/ijms20081922] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess a broad spectrum of therapeutic applications and have been used in clinical trials. MSCs are mainly retrieved from adult or fetal tissues. However, there are many obstacles with the use of tissue-derived MSCs, such as shortages of tissue sources, difficult and invasive retrieval methods, cell population heterogeneity, low purity, cell senescence, and loss of pluripotency and proliferative capacities over continuous passages. Therefore, other methods to obtain high-quality MSCs need to be developed to overcome the limitations of tissue-derived MSCs. Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are considered potent sources for the derivation of MSCs. PSC-derived MSCs (PSC-MSCs) may surpass tissue-derived MSCs in proliferation capacity, immunomodulatory activity, and in vivo therapeutic applications. In this review, we will discuss basic as well as recent protocols for the production of PSC-MSCs and their in vitro and in vivo therapeutic efficacies. A better understanding of the current advances in the production of PSC-MSCs will inspire scientists to devise more efficient differentiation methods that will be a breakthrough in the clinical application of PSC-MSCs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | | | | | | | | | | | | |
Collapse
|
7
|
Xu M, Shaw G, Murphy M, Barry F. Induced Pluripotent Stem Cell-Derived Mesenchymal Stromal Cells Are Functionally and Genetically Different From Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells 2019; 37:754-765. [PMID: 30779868 PMCID: PMC6591688 DOI: 10.1002/stem.2993] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/21/2018] [Accepted: 02/03/2019] [Indexed: 12/14/2022]
Abstract
There has been considerable interest in the generation of functional mesenchymal stromal cell (MSC) preparations from induced pluripotent stem cells (iPSCs) and this is now regarded as a potential source of unlimited, standardized, high‐quality cells for therapeutic applications in regenerative medicine. Although iMSCs meet minimal criteria for defining MSCs in terms of marker expression, there are substantial differences in terms of trilineage potential, specifically a marked reduction in chondrogenic and adipogenic propensity in iMSCs compared with bone marrow‐derived (BM) MSCs. To reveal the cellular basis underlying these differences, we conducted phenotypic, functional, and genetic comparisons between iMSCs and BM‐MSCs. We found that iMSCs express very high levels of both KDR and MSX2 compared with BM‐MSCs. In addition, BM‐MSCs had significantly higher levels of PDGFRα. These distinct gene expression profiles were maintained during culture expansion, suggesting that prepared iMSCs are more closely related to vascular progenitor cells (VPCs). Although VPCs can differentiate along the chondrogenic, osteogenic, and adipogenic pathways, they require different inductive conditions compared with BM‐MSCs. These observations suggest to us that iMSCs, based on current widely used preparation protocols, do not represent a true alternative to primary MSCs isolated from BM. Furthermore, this study highlights the fact that high levels of expression of typical MSC markers such as CD73, CD90, and CD105 are insufficient to distinguish MSCs from other mesodermal progenitors in differentiated induced pluripotent stem cell cultures. stem cells2019;37:754–765
Collapse
Affiliation(s)
- Maojia Xu
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Georgina Shaw
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Mary Murphy
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Frank Barry
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
8
|
Wang H, Li D, Zhai Z, Zhang X, Huang W, Chen X, Huang L, Liu H, Sun J, Zou Z, Fan Y, Ke Q, Lai X, Wang T, Li X, Shen H, Xiang AP, Li W. Characterization and Therapeutic Application of Mesenchymal Stem Cells with Neuromesodermal Origin from Human Pluripotent Stem Cells. Am J Cancer Res 2019; 9:1683-1697. [PMID: 31037131 PMCID: PMC6485183 DOI: 10.7150/thno.30487] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/02/2019] [Indexed: 02/06/2023] Open
Abstract
Rationale: Mesenchymal stem cells (MSC) hold great promise in the treatment of various diseases including autoimmune diseases, inflammatory diseases, etc., due to their pleiotropic properties. However, largely incongruent data were obtained from different MSC-based clinical trials, which may be partially due to functional heterogeneity among MSC. Here, we attempt to derive homogeneous mesenchymal stem cells with neuromesodermal origin from human pluripotent stem cells (hPSC) and evaluate their functional properties. Methods: Growth factors and/or small molecules were used for the differentiation of human pluripotent stem cells (hPSC) into neuromesodermal progenitors (NMP), which were then cultured in animal component-free and serum-free induction medium for the derivation and long-term expansion of MSC. The resulted NMP-MSC were detailed characterized by analyzing their surface marker expression, proliferation, migration, multipotency, immunomodulatory activity and global gene expression profile. Moreover, the in vivo therapeutic potential of NMP-MSC was detected in a mouse model of contact hypersensitivity (CHS). Results: We demonstrate that NMP-MSC express posterior HOX genes and exhibit characteristics similar to those of bone marrow MSC (BMSC), and NMP-MSC derived from different hPSC lines show high level of similarity in global gene expression profiles. More importantly, NMP-MSC display much stronger immunomodulatory activity than BMSC in vitro and in vivo, as revealed by decreased inflammatory cell infiltration and diminished production of pro-inflammatory cytokines in inflamed tissue of CHS models. Conclusion: Our results identify NMP as a new source of MSC and suggest that functional and homogeneous NMP-MSC could serve as a candidate for MSC-based therapies.
Collapse
|
9
|
Fitzsimmons REB, Mazurek MS, Soos A, Simmons CA. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem Cells Int 2018; 2018:8031718. [PMID: 30210552 PMCID: PMC6120267 DOI: 10.1155/2018/8031718] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/08/2023] Open
Abstract
As a result of over five decades of investigation, mesenchymal stromal/stem cells (MSCs) have emerged as a versatile and frequently utilized cell source in the fields of regenerative medicine and tissue engineering. In this review, we summarize the history of MSC research from the initial discovery of their multipotency to the more recent recognition of their perivascular identity in vivo and their extraordinary capacity for immunomodulation and angiogenic signaling. As well, we discuss long-standing questions regarding their developmental origins and their capacity for differentiation toward a range of cell lineages. We also highlight important considerations and potential risks involved with their isolation, ex vivo expansion, and clinical use. Overall, this review aims to serve as an overview of the breadth of research that has demonstrated the utility of MSCs in a wide range of clinical contexts and continues to unravel the mechanisms by which these cells exert their therapeutic effects.
Collapse
Affiliation(s)
- Ross E. B. Fitzsimmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Matthew S. Mazurek
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada T2N 4Z6
| | - Agnes Soos
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Craig A. Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada M5S 3G8
| |
Collapse
|
10
|
Fliefel R, Ehrenfeld M, Otto S. Induced pluripotent stem cells (iPSCs) as a new source of bone in reconstructive surgery: A systematic review and meta-analysis of preclinical studies. J Tissue Eng Regen Med 2018; 12:1780-1797. [DOI: 10.1002/term.2697] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Riham Fliefel
- Experimental Surgery and Regenerative Medicine (ExperiMed), Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry; Alexandria University; Alexandria Egypt
| | - Michael Ehrenfeld
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| | - Sven Otto
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| |
Collapse
|
11
|
Potential of iPSC-Derived Mesenchymal Stromal Cells for Treating Periodontal Disease. Stem Cells Int 2018; 2018:2601945. [PMID: 29731776 PMCID: PMC5872653 DOI: 10.1155/2018/2601945] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/31/2018] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cell-like populations have been derived from mouse-induced pluripotent stem cells (miPSC-MSC) with the capability for tissue regeneration. In this study, murine iPSC underwent differentiation towards an MSC-like immunophenotype. Stable miPSC-MSC cultures expressed the MSC-associated markers, CD73, CD105, and Sca-1, but lacked expression of the pluripotency marker, SSEA1, and hematopoietic markers, CD34 and CD45. Functionally, miPSC-MSC exhibited the potential for trilineage differentiation into osteoblasts, adipocytes, and chondrocytes and the capacity to suppress the proliferation of mitogen-activated splenocytes. The efficacy of miPSC-MSC was assessed in an acute inflammation model following systemic or local delivery into mice with subcutaneous implants containing heat-inactivated P. gingivalis. Histological analysis revealed less inflammatory cellular infiltrate within the sponges in mice treated with miPSC-MSC cells delivered locally rather than systemically. Assessment of proinflammatory cytokines in mouse spleens found that CXCL1 transcripts and protein were reduced in mice treated with miPSC-MSC. In a periodontitis model, mice subjected to oral inoculation with P. gingivalis revealed less bone tissue destruction and inflammation within the jaws when treated with miPSC-MSC compared to PBS alone. Our results demonstrated that miPSC-MSC derived from iPSC have the capacity to control acute and chronic inflammatory responses associated with the destruction of periodontal tissue. Therefore, miPSC-MSC present a promising novel source of stromal cells which could be used in the treatment of periodontal disease and other inflammatory systemic diseases such as rheumatoid arthritis.
Collapse
|
12
|
Yin X, Li P, Li Y, Cai Y, Wen J, Luan Q. Growth/differentiation factor-5 promotes in vitro/vivo periodontal specific differentiation of induced pluripotent stem cell-derived mesenchymal stem cells. Exp Ther Med 2017; 14:4111-4117. [PMID: 29067102 PMCID: PMC5647693 DOI: 10.3892/etm.2017.5030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 02/24/2017] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) derived from induced pluripotent stem cells (iPSCs) represent a promising alternative source of MSCs for effective periodontal regeneration. Scientific evidence has demonstrated that growth/differentiation factor-5 (GDF-5) supports regeneration of periodontal tissues and has a key role in MSC differentiation. The present study investigated the effects of recombinant human GDF-5 (rhGDF-5) on periodontal specific differentiation of iPSC-derived MSCs (iPSC-MSCs) and bone marrow mesenchymal stem cells (BMSCs). rhGDF-5 treatment in vitro significantly enhanced the expression levels of marker genes associated with osteogenesis (OCN), fibrogenesis (periostin) and cementogenesis (CAP) in the iPSC-MSCs compared with untreated controls (all P<0.05). Interestingly, the rhGDF-5-treated BMSCs failed to exhibit overexpression of periostin and CAP despite highly upregulated expression of OCN. In the presence of rhGDF-5, both the iPSC-MSCs and BMSCs demonstrated marked formation of mineralized nodules. Notably, rhGDF-5 greatly promoted periodontal specific differentiation of the iPSC-MSCs encapsulated in hyaluronic acid (HA) hydrogels in vivo as determined by immunohistochemical and immunofluorescence staining. The majority of the PKH67-labeled iPSC-MSCs implanted with rhGDF-5 exhibited strong expression of OCN, periostin and CAP. In conclusion, iPSC-MSCs demonstrate high periodontal specific differentiation potential in response to rhGDF-5 both in vitro and in vivo. The delivery of iPSC-MSCs and rhGDF-5 with HA hydrogel may have beneficial effects in regenerative periodontal therapy.
Collapse
Affiliation(s)
- Xiaohui Yin
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Peng Li
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China.,Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, P.R. China
| | - Yang Li
- Stem Cell Research Center and Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing 100191, P.R. China
| | - Yu Cai
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Jinhua Wen
- Stem Cell Research Center and Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing 100191, P.R. China
| | - Qingxian Luan
- Department of Periodontology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| |
Collapse
|
13
|
Can Human Embryonic Stem Cell-Derived Stromal Cells Serve a Starting Material for Myoblasts? Stem Cells Int 2017; 2017:7541734. [PMID: 28706537 PMCID: PMC5494578 DOI: 10.1155/2017/7541734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022] Open
Abstract
A large number of myocytes are necessary to treat intractable muscular disorders such as Duchenne muscular dystrophy with cell-based therapies. However, starting materials for cellular therapy products such as myoblasts, marrow stromal cells, menstrual blood-derived cells, and placenta-derived cells have a limited lifespan and cease to proliferate in vitro. From the viewpoints of manufacturing and quality control, cells with a long lifespan are more suitable as a starting material. In this study, we generated stromal cells for future myoblast therapy from a working cell bank of human embryonic stem cells (ESCs). The ESC-derived CD105+ cells with extensive in vitro proliferation capability exhibited myogenesis and genetic stability in vitro. These results imply that ESC-derived CD105+ cells are another cell source for myoblasts in cell-based therapy for patients with genetic muscular disorders. Since ESCs are immortal, mesenchymal stromal cells generated from ESCs can be manufactured at a large scale in one lot for pharmaceutical purposes.
Collapse
|
14
|
Nan W, Xu Z, Chen Z, Yuan X, Lin J, Feng H, Lian J, Chen H. Bone marrow mesenchymal stem cells accelerate the hyperglycemic refractory wound healing by inhibiting an excessive inflammatory response. Mol Med Rep 2017; 15:3239-3244. [DOI: 10.3892/mmr.2017.6400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/17/2017] [Indexed: 11/05/2022] Open
|
15
|
Mesenchymal Stem and Progenitor Cells in Regeneration: Tissue Specificity and Regenerative Potential. Stem Cells Int 2017; 2017:5173732. [PMID: 28286525 PMCID: PMC5327785 DOI: 10.1155/2017/5173732] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/07/2016] [Indexed: 12/15/2022] Open
Abstract
It has always been an ambitious goal in medicine to repair or replace morbid tissues for regaining the organ functionality. This challenge has recently gained momentum through considerable progress in understanding the biological concept of the regenerative potential of stem cells. Routine therapeutic procedures are about to shift towards the use of biological and molecular armamentarium. The potential use of embryonic stem cells and invention of induced pluripotent stem cells raised hope for clinical regenerative purposes; however, the use of these interventions for regenerative therapy showed its dark side, as many health concerns and ethical issues arose in terms of using these cells in clinical applications. In this regard, adult stem cells climbed up to the top list of regenerative tools and mesenchymal stem cells (MSC) showed promise for regenerative cell therapy with a rather limited level of risk. MSC have been successfully isolated from various human tissues and they have been shown to offer the possibility to establish novel therapeutic interventions for a variety of hard-to-noncurable diseases. There have been many elegant studies investigating the impact of MSC in regenerative medicine. This review provides compact information on the role of stem cells, in particular, MSC in regeneration.
Collapse
|
16
|
Kindlin-2 Modulates the Survival, Differentiation, and Migration of Induced Pluripotent Cell-Derived Mesenchymal Stromal Cells. Stem Cells Int 2017; 2017:7316354. [PMID: 28163724 PMCID: PMC5253493 DOI: 10.1155/2017/7316354] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/24/2016] [Accepted: 12/12/2016] [Indexed: 01/21/2023] Open
Abstract
Kindlin-2 is a multidomain intracellular protein that can be recruited to β-integrin domains to activate signaling, initiate transcriptional programs, and bind to E-cadherin. To explore its involvement in cell fate decisions in mesenchymal cells, we studied the effects of Kindlin-2 modification (overexpression/knockdown) in induced pluripotent cell-derived mesenchymal stromal cells (iPSC-MSCs). Kindlin-2 overexpression resulted in increased proliferation and reduced apoptosis of iPSC-MSCs, as well as inhibition of their differentiation towards osteocytes, adipocytes, and chondrocytes. In contrast, siRNA-mediated Kindlin-2 knockdown induced increased apoptosis and increased differentiation response in iPSC-MSCs. The ability of iPSC-MSCs to adhere to VCAM-1/SDF-1α under shear stress and to migrate in a wound scratch assay was significantly increased after Kindlin-2 overexpression. In contrast, inhibition of mixed lymphocyte reaction (MLR) was generally independent of Kindlin-2 modulation in iPSC-MSCs, except for decreased production of interleukin-2 (IL-2) after Kindlin-2 overexpression in iPS-MSCs. Thus, Kindlin-2 upregulates survival, proliferation, stemness, and migration potential in iPSC-MSCs and may therefore be beneficial in optimizing performance of iPSC-MSC in therapies.
Collapse
|
17
|
Vasko T, Frobel J, Lubberich R, Goecke TW, Wagner W. iPSC-derived mesenchymal stromal cells are less supportive than primary MSCs for co-culture of hematopoietic progenitor cells. J Hematol Oncol 2016; 9:43. [PMID: 27098268 PMCID: PMC4839158 DOI: 10.1186/s13045-016-0273-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/13/2016] [Indexed: 02/11/2023] Open
Abstract
In vitro culture of hematopoietic stem and progenitor cells (HPCs) is supported by a suitable cellular microenvironment, such as mesenchymal stromal cells (MSCs)-but MSCs are heterogeneous and poorly defined. In this study, we analyzed whether MSCs derived from induced pluripotent stem cells (iPS-MSCs) provide a suitable cellular feeder layer too. iPS-MSCs clearly supported proliferation of HPCs, maintenance of a primitive immunophenotype (CD34(+), CD133(+), CD38(-)), and colony-forming unit (CFU) potential of CD34(+) HPCs. However, particularly long-term culture-initiating cell (LTC-IC) frequency was lower with iPS-MSCs as compared to primary MSCs. Relevant genes for cell-cell interaction were overall expressed at similar level in MSCs and iPS-MSCs, whereas VCAM1 was less expressed in the latter. In conclusion, our iPS-MSCs support in vitro culture of HPCs; however, under the current differentiation and culture conditions, they are less suitable than primary MSCs from bone marrow.
Collapse
Affiliation(s)
- Theresa Vasko
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany.,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Joana Frobel
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany.,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Richard Lubberich
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany.,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Tamme W Goecke
- Department of Obstetrics and Gynecology, RWTH Aachen University Hospital, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Pauwelsstrasse 20, 52074, Aachen, Germany. .,Institute for Biomedical Engineering-Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|