1
|
Chen S, Liang B, Xu J. Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations. J Transl Med 2024; 22:459. [PMID: 38750573 PMCID: PMC11094970 DOI: 10.1186/s12967-024-05294-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/11/2024] [Indexed: 05/19/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a heterogeneous cell population distributed throughout various tissues, demonstrating remarkable adaptability to microenvironmental cues and holding immense promise for disease treatment. However, the inherent diversity within MSCs often leads to variability in therapeutic outcomes, posing challenges for clinical applications. To address this heterogeneity, purification of MSC subpopulations through marker-based isolation has emerged as a promising approach to ensure consistent therapeutic efficacy. In this review, we discussed the reported markers of MSCs, encompassing those developed through candidate marker strategies and high-throughput approaches, with the aim of explore viable strategies for addressing the heterogeneity of MSCs and illuminate prospective research directions in this field.
Collapse
Affiliation(s)
- Si Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Bowei Liang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Jianyong Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Fuqiang Avenue 1001, Shenzhen, 518060, Guangdong, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
| |
Collapse
|
2
|
Song J, Ma Q, Li Y, Wang X, Chen S, Liang B, Lin X, Chen J, Xu S, Shi S, Zhang J, Diao L, Zeng Y, Xu J. CD317 + MSCs expanded with chemically defined media have enhanced immunological anti-inflammatory activities. Stem Cell Res Ther 2024; 15:2. [PMID: 38169422 PMCID: PMC10763464 DOI: 10.1186/s13287-023-03618-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Although both preclinical and clinical studies have shown the great application potential of MSCs (mesenchymal stem/stromal cells) in treating many kinds of diseases, therapeutic inconsistency resulting from cell heterogeneity is the major stumbling block to their clinical applications. Cell population diversity and batch variation in the cell expansion medium are two major inducers of MSC heterogeneity. METHODS Cell population diversity was investigated through single-cell RNA sequencing analysis of human MSCs derived from the umbilical cord and expanded with fully chemically defined medium in the current study. Then, the MSC subpopulation with enhanced anti-inflammatory effects was studied in vitro and in vivo. RESULTS Our data showed that MSCs contain different populations with different functions, including subpopulations with enhanced functions of exosome secretion, extracellular matrix modification and responses to stimuli (regeneration and immune response). Among them, CD317+ MSCs have improved differentiation capabilities and enhanced immune suppression activities. Underlying mechanism studies showed that higher levels of TSG6 confer enhanced anti-inflammatory functions of CD317+ MSCs. CONCLUSIONS Thus, CD317+ MSCs might be a promising candidate for treating immunological disorder-related diseases.
Collapse
Affiliation(s)
- Jun Song
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150000, People's Republic of China
| | - Qi Ma
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150000, People's Republic of China
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Yumeng Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150000, People's Republic of China
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Xianqi Wang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Si Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Bowei Liang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Xiaoqi Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Jieting Chen
- Department of Obstetrics, People's Hospital of Baoan, Shenzhen, 518000, People's Republic of China
| | - Shiru Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Shaoquan Shi
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Jingting Zhang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Yong Zeng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Jianyong Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Fuqiang Avenue 1001, Shenzhen, 518060, Guangdong, People's Republic of China.
| |
Collapse
|
3
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
4
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
5
|
Nowlan B, Williams ED, Doran MR. Direct bone marrow injection of human bone marrow-derived stromal cells into mouse femurs results in greater prostate cancer PC-3 cell proliferation, but not specifically proliferation within the injected femurs. BMC Cancer 2022; 22:554. [PMID: 35581599 PMCID: PMC9112579 DOI: 10.1186/s12885-022-09430-6] [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: 10/13/2021] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
Background While prostate cancer (PCa) cells most often metastasize to bone in men, species-specific differences between human and mouse bone marrow mean that this pattern is not faithfully replicated in mice. Herein we evaluated the impact of partially humanizing mouse bone marrow with human bone marrow-derived stromal cells (BMSC, also known as "mesenchymal stem cells") on human PCa cell behaviour. Methods BMSC are key cellular constituents of marrow. We used intrafemoral injection to transplant 5 × 105 luciferase (Luc) and green fluorescence protein (GFP) expressing human BMSC (hBMSC-Luc/GFP) into the right femur of non-obese diabetic (NOD)-severe combined immunodeficiency (scid) interleukin (IL)-2γ−/− (NSG) mice. Two weeks later, 2.5 × 106 PC-3 prostate cancer cells expressing DsRed (PC-3-DsRed) were delivered into the mice via intracardiac injection. PC-3-DsRed cells were tracked over time using an In Vivo Imaging System (IVIS) live animal imaging system, X-ray and IVIS imaging performed on harvested organs, and PC-3 cell numbers in femurs quantified using flow cytometry and histology. Results Flow cytometry analysis revealed greater PC-3-DsRed cell numbers within femurs of the mice that received hBMSC-Luc/GFP. However, while there were overall greater PC-3-DsRed cell numbers in these animals, there were not more PC-3-DsRed in the femurs injected with hBMSC-Luc/GFP than in contralateral femurs. A similar proportion of mice in with or without hBMSC-Luc/GFP had bone lessions, but the absolute number of bone lesions was greater in mice that had received hBMSC-Luc/GFP. Conclusion PC-3-DsRed cells preferentially populated bones in mice that had received hBMSC-Luc/GFP, although PC-3-DsRed cells not specifically localize in the bone marrow cavity where hBMSC-Luc/GFP had been transplanted. hBMSC-Luc/GFP appear to modify mouse biology in a manner that supports PC-3-DsRed tumor development, rather than specifically influencing PC-3-DsRed cell homing. This study provides useful insights into the role of humanizing murine bone marrow with hBMSC to study human PCa cell biology. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09430-6.
Collapse
Affiliation(s)
- Bianca Nowlan
- School of Biomedical Science, Faculty of Health, Queensland University of Technology at the Translational Research Institute, Brisbane, Australia.,Australian Prostate Cancer Research Centre-Queensland, Brisbane, Australia
| | - Elizabeth D Williams
- School of Biomedical Science, Faculty of Health, Queensland University of Technology at the Translational Research Institute, Brisbane, Australia.,Australian Prostate Cancer Research Centre-Queensland, Brisbane, Australia
| | - Michael Robert Doran
- School of Biomedical Science, Faculty of Health, Queensland University of Technology at the Translational Research Institute, Brisbane, Australia. .,Australian Prostate Cancer Research Centre-Queensland, Brisbane, Australia. .,Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia. .,Mater Research Institute - University of Queensland, Brisbane, Australia. .,Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
6
|
Cabanillas Stanchi KM, Böhringer J, Strölin M, Groeschel S, Lenglinger K, Treuner C, Kehrer C, Laugwitz L, Bevot A, Kaiser N, Schumm M, Lang P, Handgretinger R, Krägeloh-Mann I, Müller I, Döring M. Hematopoietic stem cell transplantation with mesenchymal stromal cells in children with metachromatic leukodystrophy. Stem Cells Dev 2022; 31:163-175. [PMID: 35323019 DOI: 10.1089/scd.2021.0352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disorder primarily affecting the white matter of the nervous system that results from a deficiency of the arylsulfatase A (ARSA). Mesenchymal stem cells (MSCs) are able to secrete ARSA and have shown beneficial effects in MLD patients. In this retrospective analysis, 10 pediatric MLD patients (MSCG) underwent allogeneic hematopoietic stem cell transplantation (HSCT) and received two applications of 2 x 106 MSCs/kg bodyweight at day +30 and +60 after HSCT between 2007 and 2018. MSC safety, occurrence of graft-versus-host disease (GvHD), blood ARSA levels, chimerism, cell regeneration and engraftment, MRI changes, and the gross motor function were assessed within the first year of HSCT. The long-term data included clinical outcomes and safety aspects of MSCs. Data were compared to a control cohort of seven pediatric MLD patients (CG) who underwent HSCT only. The application of MSC in pediatric MLD patients after allogeneic HSCT was safe and well tolerated and long-term potentially MSC-related adverse effects up to 13.5 years after HSCT were not observed. Patients achieved significantly higher ARSA levels (CG: median 1.03 nmol∙10-6, range 0.41-1.73 | MSCG: median 1.58 nmol∙10-6, range 0.44-2.6; p<0.05), as well as significantly higher leukocyte (p<0.05) and thrombocyte (p<0.001) levels within 365 days of MSC application compared to CG patients. Statistically significant effects on acute GvHD, regeneration of immune cells, engraftment, MRI changes, gross motor function, and clinical outcomes were not detected. In conclusion, the application of MSCs in pediatric MLD patients after allogeneic HSCT was safe and well tolerated. The two applications of 2 x 106/kg allogeneic MSCs were followed by improved engraftment and hematopoiesis within the first year after HSCT. Larger, prospective trials are necessary to evaluate the impact of MSC application on engraftment and hematopoietic recovery.
Collapse
Affiliation(s)
| | - Judith Böhringer
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Manuel Strölin
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Samuel Groeschel
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Katrin Lenglinger
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Claudia Treuner
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Christiane Kehrer
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Lucia Laugwitz
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Andrea Bevot
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Nadja Kaiser
- University Children's Hospital Tübingen, Dpt. III - Neuropediatrics, Germany;
| | - Michael Schumm
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Peter Lang
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| | - Rupert Handgretinger
- Children's University Hospital, Hematology/Oncology, Hoppe-Seyler-Str. 1, Tuebingen, Germany, 72076;
| | | | - Ingo Müller
- University Medical Center Hamburg-Eppendorf, 37734, Department of Pediatric Hematology and Oncology, Hamburg, Hamburg, Germany;
| | - Michaela Döring
- University Children's Hospital Tübingen, Dpt. I - General Pediatrics, Hematology and Oncology, Germany;
| |
Collapse
|
7
|
Ji X, Sun T, Xie S, Qian H, Song L, Wang L, Liu H, Feng Q. Upregulation of CPNE7 in mesenchymal stromal cells promotes oral squamous cell carcinoma metastasis through the NF-κB pathway. Cell Death Discov 2021; 7:294. [PMID: 34650058 PMCID: PMC8516970 DOI: 10.1038/s41420-021-00684-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
A remarkable shift in Mesenchymal stromal cells (MSCs) plays an important role in cancer metastasis, but the molecular mechanism is still unclear. CPNE7, a calcium-dependent phospholipid-binding protein, mediates signal transduction and metastasis in many tumours. Here, we demonstrated that MSCs derived from OSCC (OSCC-MSCs) promoted the metastasis of OSCC cells by transwell assay and animal models through epithelial to mesenchymal transition (EMT) (p < 0.05). RNA-sequencing, ELISA, neutralizing antibody and CXCR2 inhibitor assay confirmed that CXCL8 secreted by OSCC-MSCs was associated with the upregulated expression of CPNE7 by immunohistochemical and western blotting (p < 0.05). This is mechanistically linked to the activation of CPNE7 to NF-κB pathway-induced metastasis, including phosphorylated p65 and IκBa. CPNE7 silencing inhibited metastatic abilities and the expression of CXCL8, phosphorylated p65, IκBa, and p65 nuclear translocation by western blotting and immunofluorescence, while CPNE7 overexpression markedly promoted these events (p < 0.05). We also identified that Nucleolin could be bind CPNE7 and IκBa by co-immunoprecipitation. Together, our results suggest that upregulation of CPNE7 in MSCs interacted with surface receptor -Nucleolin and then combined with IκBa to promoted phosphorylated IκBa and p65 nuclear translocation to active NF-κB pathway, and then regulates CXCL8 secretion to promote the metastasis of OSCC cells. Therefore, CPNE7 in MSCs could be promising therapeutic targets in OSCC.
Collapse
Affiliation(s)
- Xiaoli Ji
- Department of Stomatology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No.105 Jiefang Road, Jinan, 250013, Shandong, China. .,Department of Oral Mucosal Diseases, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, 250012, Shandong, China. .,Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, 250012, Shandong, China.
| | - Tianyong Sun
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, 250012, Shandong, China
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian, Beijing, 100081, China
| | - Hua Qian
- Department of Stomatology, The Second Hospital of Shandong University, No. 247 Beiyuan Road, Jinan, 250033, China
| | - Lixiang Song
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, 250012, Shandong, China
| | - Lihua Wang
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, 250012, Shandong, China
| | - Hongwei Liu
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian, Beijing, 100081, China.
| | - Qiang Feng
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Provincial Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, 250012, Shandong, China. .,NHC Key Laboratory of Otorhinolaryngology (Shandong University), No.44-1 Wenhua Road West, Jinan, Shandong, China, 250012.
| |
Collapse
|
8
|
Doron G, Temenoff JS. Culture Substrates for Improved Manufacture of Mesenchymal Stromal Cell Therapies. Adv Healthc Mater 2021; 10:e2100016. [PMID: 33930252 DOI: 10.1002/adhm.202100016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Recent developments in mesenchymal stromal cell (MSC) therapies have increased the demand for tools to improve their manufacture, including the selection of optimal culture substrate materials. While many clinical manufacturers use planar tissue culture plastic (TCP) surfaces for MSC production, others have begun exploring the use of alternative culture substrates that present a variety of spatial, mechanical, and biochemical cues that influence cell expansion and resulting cell quality. In this review, the effects of culture and material properties distinct from traditional planar TCP surfaces on MSC proliferation, surface marker expression, and commonly used indications for therapeutic potency are examined. The different properties summarized include the use of alternative culture formats such as cellular aggregates or 3D scaffolds, as well as the effects of culture substrate stiffness and presentation of specific adhesive ligands and topographical cues. Specific substrate properties can be related to greater cell expansion and improvement in specific therapeutic functionalities, demonstrating the utility of culture materials in further improving the clinical-scale manufacture of highly secretory MSC products.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University 313 Ferst Drive Atlanta GA 30332 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology Atlanta GA 30332 USA
| | - Johnna S. Temenoff
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University 313 Ferst Drive Atlanta GA 30332 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology Atlanta GA 30332 USA
| |
Collapse
|
9
|
Diallo AM, Rota S, Boissière M, Bardonnet R, Pauthe E, Petite H, Benoist HM, Bensidhoum M, Anagnostou F. Osteoformation potential of an allogenic partially demineralized bone matrix in critical-size defects in the rat calvarium. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112207. [PMID: 34225859 DOI: 10.1016/j.msec.2021.112207] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 11/27/2022]
Abstract
Allogenic demineralized bone matrix has been developed as a reliable alternative to the autologous bone graft. In the present study, we assessed the osteoformation potential of a partially demineralized bone matrix (PDBM) in a paste form obtained without an added carrier. This formulation included the preparation of cancelous bone from femoral heads after decellularision, delipidation, demineralization in HCl and autoclaving at 121 °C. Structural and biochemical characteristics of PDBM were determined using FTIR (Fourier transform infrared spectroscopy), hydroxyproline, DNA content assays, and optical ellipsometry. The osteoformation potential was evaluated in 8-, 6-, and 4-mm-diameter rat-calvarial bone defects by in vivo micro-CT analysis, performed immediately after surgery on days 0, 15, 30, 45, and 60. Moreover, histological and histomorphometric analyses were done on day 60. PDBM was compared to cancelous bone powder (BP) before its partial demineralization. The expression levels of selected inflammation-, angiogenesis-, and bone-related genes were also investigated by RT-PCR, 3, 7, and 14 days after surgery. Compared to the control group, the PDBM group exhibited a significant increase (p < 0.05) in radiopacity in 8-mm- and 6-mm-diameter defects at all time points tested. On day 60, the amount of newly-formed bone was greater (16 and 1.6 folds; p < 0.001; respectively) compared to that in control defects. No bone formation was observed in defects filled with BP regardeless of the size. In 8-mm-diameter defect, PDBM was effective enough to induce the upregulation of genes pertinent to inflammation (i.e., TNFα, IL-6, and IL-8), angiogenesis (i.e., VEGF, VWF), and osteogenesis (ALP, RUNX2, BGLAP, SP7) by day 3 after surgery. This study showed that the tested PDBM deeply influences the early critical events involved in bone regeneration and exhibits efficient osteoformation capacity, making it an attractive graft option for treating defects in periodontal and maxillofacial areas.
Collapse
Affiliation(s)
- Ahmad Moustapha Diallo
- CNRS, UMR 7052 - INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Universiy of Paris, 10 Avenue de Verdun, 75010 Paris, France; Service of Periodontology, Institute of Odontology and Stomatology (IOS), University Cheikh Anta Diop (UCAD), BP 5005 Dakar-Fann, Sénégal; Faculty of Medecine, Pharmacy and Odonto-Stomatology, University Cheikh Anta Diop (UCAD), BP 5005 Dakar-Fann, Sénégal
| | - Solène Rota
- ERRMECe, Research Team on Extracellular Matrix-Cellular Relationships (EA1391), Biomaterials for Health Research Group, Institute of Materials I-MAT (FD4122), CY Tech, CY University Cergy Paris, International House of Research (MIR), rue Descartes, 95001 Neuville sur Oise cedex, France; Biobank, 3 rue Georges Charpak, 77127 Lieusaint, France
| | - Michel Boissière
- ERRMECe, Research Team on Extracellular Matrix-Cellular Relationships (EA1391), Biomaterials for Health Research Group, Institute of Materials I-MAT (FD4122), CY Tech, CY University Cergy Paris, International House of Research (MIR), rue Descartes, 95001 Neuville sur Oise cedex, France
| | | | - Emmanuel Pauthe
- ERRMECe, Research Team on Extracellular Matrix-Cellular Relationships (EA1391), Biomaterials for Health Research Group, Institute of Materials I-MAT (FD4122), CY Tech, CY University Cergy Paris, International House of Research (MIR), rue Descartes, 95001 Neuville sur Oise cedex, France
| | - Hervé Petite
- CNRS, UMR 7052 - INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Universiy of Paris, 10 Avenue de Verdun, 75010 Paris, France
| | - Henri M Benoist
- Service of Periodontology, Institute of Odontology and Stomatology (IOS), University Cheikh Anta Diop (UCAD), BP 5005 Dakar-Fann, Sénégal; Faculty of Medecine, Pharmacy and Odonto-Stomatology, University Cheikh Anta Diop (UCAD), BP 5005 Dakar-Fann, Sénégal
| | - Morad Bensidhoum
- CNRS, UMR 7052 - INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Universiy of Paris, 10 Avenue de Verdun, 75010 Paris, France
| | - Fani Anagnostou
- CNRS, UMR 7052 - INSERM U1271, Laboratory of Osteoarticular Biology, Bioengineering and Bioimaging, Universiy of Paris, 10 Avenue de Verdun, 75010 Paris, France; Service of Odontology, Hôpital Pitié-Salpêtrière APHP, U.F.R. of Odontology University of Paris, 47-83 Boulevard de l'Hôpital, 75013 Paris, France.
| |
Collapse
|
10
|
Nowlan B, Futrega K, Williams ED, Doran MR. Human bone marrow-derived stromal cell behavior when injected directly into the bone marrow of NOD-scid-gamma mice pre-conditioned with sub-lethal irradiation. Stem Cell Res Ther 2021; 12:231. [PMID: 33845908 PMCID: PMC8042930 DOI: 10.1186/s13287-021-02297-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Direct bone marrow injection of cells into murine marrow cavities is used in a range of cell characterization assays and to develop disease models. While human bone marrow-derived stromal cells (hBMSC, also known as mesenchymal stem cells (MSC)) are frequently described in therapeutic applications, or disease modeling, their behavior following direct injection into murine bone marrow is poorly characterized. Herein, we characterized hBMSC engraftment and persistence within the bone marrow of NOD-scid interleukin (IL)-2γ-/- (NSG) mice with or without prior 2 Gy total-body γ-irradiation of recipient mice. METHODS One day after conditioning NSG mice with sublethal irradiation, 5 × 105 luciferase (Luc) and green fluorescent protein (GFP)-expressing hBMSC (hBMSC-Luc/GFP) were injected into the right femurs of animals. hBMSC-Luc/GFP were tracked in live animals using IVIS imaging, and histology was used to further characterize hBMSC location and behavior in tissues. RESULTS hBMSC-Luc/GFP number within injected marrow cavities declined rapidly over 4 weeks, but prior irradiation of animals delayed this decline. At 4 weeks, hBMSC-Luc/GFP colonized injected marrow cavities and distal marrow cavities at rates of 2.5 ± 2.2% and 1.7 ± 1.9% of total marrow nucleated cells, respectively in both irradiated and non-irradiated mice. In distal marrow cavities, hBMSC were not uniformly distributed and appeared to be co-localized in clusters, with the majority found in the endosteal region. CONCLUSIONS While significant numbers of hBMSC-Luc/GFP could be deposited into the mouse bone marrow via direct bone marrow injection, IVIS imaging indicated that the number of hBMSC-Luc/GFP in that bone marrow cavity declined with time. Irradiation of mice prior to transplant only delayed the rate of hBMSC-Luc/GFP population decline in injected femurs. Clusters of hBMSC-Luc/GFP were observed in the histology of distal marrow cavities, suggesting that some transplanted cells actively homed to distal marrow cavities. Individual cell clusters may have arisen from discrete clones that homed to the marrow, and then underwent modest proliferation. The transient high-density population of hBMSC within the injected femur, or the longer-term low-density population of hBMSC in distal marrow cavities, offers useful models for studying disease or regenerative processes. Experimental designs should consider how relative hBMSC distribution and local hBMSC densities evolve over time.
Collapse
Affiliation(s)
- Bianca Nowlan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, Queensland, Australia.,Translational Research Institute, 37 Kent Street, Brisbane, Queensland, 4102, Australia
| | - Kathryn Futrega
- Translational Research Institute, 37 Kent Street, Brisbane, Queensland, 4102, Australia.,Centre for Biomedical Technologies (CBT) and School of Mechanical, Medical, and Process Engineering (MMPE), Queensland University of Technology (QUT), Brisbane, Australia.,Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, 30 Convent Dr MSC 4320, Bethesda, MD, 20892-4320, USA
| | - Elizabeth Deborah Williams
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, Queensland, Australia.,Translational Research Institute, 37 Kent Street, Brisbane, Queensland, 4102, Australia
| | - Michael Robert Doran
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Australia. .,Australian Prostate Cancer Research Centre - Queensland (APCCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, Queensland, Australia. .,Translational Research Institute, 37 Kent Street, Brisbane, Queensland, 4102, Australia. .,Centre for Biomedical Technologies (CBT) and School of Mechanical, Medical, and Process Engineering (MMPE), Queensland University of Technology (QUT), Brisbane, Australia. .,Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, 30 Convent Dr MSC 4320, Bethesda, MD, 20892-4320, USA. .,Mater Research Institute - University of Queensland, Brisbane, Australia. .,Australian National Centre for the Public Awareness of Science, Australian National University, Canberra, Australia.
| |
Collapse
|
11
|
To Breathe or Not to Breathe: The Role of Oxygen in Bone Marrow-Derived Mesenchymal Stromal Cell Senescence. Stem Cells Int 2021; 2021:8899756. [PMID: 33519938 PMCID: PMC7817290 DOI: 10.1155/2021/8899756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/09/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based cellular therapy is a promising tool for the treatment of pathological conditions with underlying severe tissue damage or malfunction like in chronic cardiovascular, musculoskeletal, or inflammatory conditions. One of the biggest technical challenges of the use of natural stem cells, however, is the prevention of their premature senescence during therapeutical manipulations. Culturing stem cells under hypoxic conditions is believed to be a possible route to fulfill this goal. Here, we review current literature data on the effects of hypoxia on bone marrow-derived mesenchymal stromal cells, one of the most popular tools of practical cellular therapy, in the context of their senescence.
Collapse
|
12
|
Couto de Carvalho LA, Tosta Dos Santos SL, Sacramento LV, de Almeida VR, de Aquino Xavier FC, Dos Santos JN, Gomes Henriques Leitão ÁC. Mesenchymal stem cell markers in periodontal tissues and periapical lesions. Acta Histochem 2020; 122:151636. [PMID: 33132168 DOI: 10.1016/j.acthis.2020.151636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) are characterized by the potential to differentiate into multiple cell lineages, high proliferation rates, and self-renewal capacity, in addition to the ability to maintain their undifferentiated state. These cells have been identified in physiological oral tissues such as pulp tissue, dental follicle, apical papilla and periodontal ligament, as well as in pathological situations such as chronic periapical lesions (CPLs). The criteria used for the identification of MSCs include the positive expression of specific surface antigens, with CD73, CD90, CD105, CD44, CD146, STRO-1, CD166, NANOG and OCT4 being the most specific for these cells. AIM The aim of this review was to explore the literature on markers able to identify MSCs as well as the presence of these cells in the healthy periodontal ligament and CPLs, highlighting their role in regenerative medicine and implications in the progression of these lesions. METHODS Narrative literature review searching the PubMed and Medline databases. Articles published in English between 1974 and 2020 were retrieved. CONCLUSION The included studies confirmed the presence of MSCs in the healthy periodontal ligament and in CPLs. Several surface markers are used for the characterization of these cells which, although not specific, are effective in cell recognition. Mesenchymal stem cells participate in tissue repair, exerting anti- inflammatory, immunosuppressive and proangiogenic effects, and are therefore involved in the progression and attenuation of CPLs or even in the persistence of these lesions.
Collapse
Affiliation(s)
| | | | | | | | | | - Jean Nunes Dos Santos
- Postgraduation Program in Dentistry and Health, Federal University of Bahia, Salvador, BA, Brazil
| | | |
Collapse
|
13
|
Abstract
Over the last decade, there has been a considerable progress in the development of cell therapy products for the treatment of liver diseases. The quest to generate well-defined homogenous cell populations with defined mechanism(s) of action has enabled the progression from use of autologous bone marrow stem cells comprising of heterogeneous cell populations to allogeneic cell types such as monocyte-derived macrophages, regulatory T cells, mesenchymal stromal cells, macrophages, etc. There is growing evidence regarding the multiple molecular mechanisms pivotal to various therapeutic effects and hence, careful selection of cell therapy product for the desired putative effects is crucial. In this review, we have presented an overview of the cell therapies that have been developed thus far, with preclinical and clinical evidence for their use in liver disease. Limitations associated with these therapies have also been discussed. Despite the advances made, there remain multiple challenges to overcome before cell therapies can be considered as viable treatment options, and these include larger scale clinical trials, scalable production of cells according to good manufacturing practice standards, pathways for delivery of cell therapy within hospital environments, and costs associated with the production.
Collapse
Affiliation(s)
- Sheeba Khan
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Reenam S Khan
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
14
|
Liu Z, Wu X, Wang S, Xia L, Xiao H, Li Y, Li H, Zhang Y, Xu D, Nie D, Lai Y, Wu B, Lin D, Du X, Jiang Z, Gao Y, Gu X, Xiao Y. Co-transplantation of mesenchymal stem cells makes haploidentical HSCT a potential comparable therapy with matched sibling donor HSCT for patients with severe aplastic anemia. Ther Adv Hematol 2020; 11:2040620720965411. [PMID: 33194162 PMCID: PMC7605036 DOI: 10.1177/2040620720965411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
The application of haploidentical hematopoietic stem cell transplantation (HSCT) with mesenchymal stem cell (MSC) infusion as a treatment regimen for severe aplastic anemia (SAA) has been reported to be efficacious in single-arm trials. However, it is difficult to assess without comparing the results with those from a first-line, matched-sibling HSCT. Herein, we retrospectively reviewed 91 patients with acquired SAA. They received HSCT from haploidentical donors combined with MSC transfer (HID group). We compared these patients with 103 others who received first-line matched-sibling HSCT (MSD group) to evaluate relative treatment efficacy. Compared with the patients in the MSD group, those in the HID group presented with higher incidences of grades II–IV and III–IV acute graft versus host disease (aGvHD) and chronic graft versus host disease (cGvHD) (p < 0.05). However, the incidence of myeloid and platelet engraftment, graft failure, poor graft function, and extensive cGvHD were comparable for both groups. The median follow-up was 36.6 months and the 3-year overall survival rate was similar for both groups (83.5% versus 79.1%). Univariate and multivariate analyses revealed that time intervals greater than 4 months from diagnosis to transplantation, experienced graft failure, poor graft function, or grade III–IV aGvHD were significantly associated with adverse outcomes. All HID patients received MSC co-transplantation with hematopoietic stem cells. However, the infused MSCs were derived from umbilical cord (UC-MSC group; 43 patients) or bone marrow (BM-MSC group; 48 patients) and were administered at different medical centers. We first compared the outcomes between the two groups and detected that the BM-MSC group exhibited lower incidences of grade III–IV aGvHD and cGvHD (p < 0.05). This study suggests that co-transplantation of hematopoietic and MSCs significantly reduces the risk and incidence of graft rejection and may effectively improve overall survival in patients with SAA even in the absence of closely related histocompatible donor material.
Collapse
Affiliation(s)
- Zenghui Liu
- Guangzhou University of Chinese Medicine; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoxiong Wu
- First Affiliated Hospital of PLA General Hospital, Beijing, China
| | | | - Linghui Xia
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haowen Xiao
- General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yonghua Li
- General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Hongbo Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuping Zhang
- Guangzhou First People's Hospital, Guangzhou, China
| | - Duorong Xu
- First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Danian Nie
- Second Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yongrong Lai
- First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bingyi Wu
- Affiliated Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Dongjun Lin
- Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xin Du
- Shenzhen Second People's Hospital, Shenzhen, China
| | - Zujun Jiang
- General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Yang Gao
- General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Xuekui Gu
- Department of Hematology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No 16, Jichang Road, Guangzhou, Guangdong Province, 510405, PR China
| | - Yang Xiao
- Stem Cell Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, No. 250, Changgang East Road, Guangzhou, Guangdong Province, 510260, PR China
| |
Collapse
|
15
|
Childs PG, Reid S, Salmeron-Sanchez M, Dalby MJ. Hurdles to uptake of mesenchymal stem cells and their progenitors in therapeutic products. Biochem J 2020; 477:3349-3366. [PMID: 32941644 PMCID: PMC7505558 DOI: 10.1042/bcj20190382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022]
Abstract
Twenty-five years have passed since the first clinical trial utilising mesenchymal stomal/stem cells (MSCs) in 1995. In this time academic research has grown our understanding of MSC biochemistry and our ability to manipulate these cells in vitro using chemical, biomaterial, and mechanical methods. Research has been emboldened by the promise that MSCs can treat illness and repair damaged tissues through their capacity for immunomodulation and differentiation. Since 1995, 31 therapeutic products containing MSCs and/or progenitors have reached the market with the level of in vitro manipulation varying significantly. In this review, we summarise existing therapeutic products containing MSCs or mesenchymal progenitor cells and examine the challenges faced when developing new therapeutic products. Successful progression to clinical trial, and ultimately market, requires a thorough understanding of these hurdles at the earliest stages of in vitro pre-clinical development. It is beneficial to understand the health economic benefit for a new product and the reimbursement potential within various healthcare systems. Pre-clinical studies should be selected to demonstrate efficacy and safety for the specific clinical indication in humans, to avoid duplication of effort and minimise animal usage. Early consideration should also be given to manufacturing: how cell manipulation methods will integrate into highly controlled workflows and how they will be scaled up to produce clinically relevant quantities of cells. Finally, we summarise the main regulatory pathways for these clinical products, which can help shape early therapeutic design and testing.
Collapse
Affiliation(s)
- Peter G. Childs
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, U.K
- Centre for the Cellular Microenvironment, SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, U.K
| | - Stuart Reid
- Centre for the Cellular Microenvironment, SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, U.K
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Matthew J. Dalby
- Centre for the Cellular Microenvironment, Institute for Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| |
Collapse
|
16
|
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
|
17
|
Raza SS, Khan MA. Mesenchymal Stem Cells: A new front emerge in COVID19 treatment. Cytotherapy 2020; 24:755-766. [PMID: 35880307 PMCID: PMC7362822 DOI: 10.1016/j.jcyt.2020.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/08/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Currently, treating coronavirus disease 2019 (COVID-19) patients, particularly those afflicted with severe pneumonia, is challenging, as no effective pharmacotherapy for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exists. Severe pneumonia is recognized as a clinical syndrome characterized by hyper-induction of pro-inflammatory cytokine production, which can induce organ damage, followed by edema, dysfunction of air exchange, acute respiratory distress syndrome, acute cardiac injury, secondary infection and increased mortality. Owing to the immunoregulatory and differentiation potential of mesenchymal stem cells (MSCs), we aimed to outline current insights into the clinical application of MSCs in COVID-19 patients. Based on results from preliminary clinical investigations, it can be predicted that MSC therapy for patients infected with SARS-CoV-2 is safe and effective, although multiple clinical trials with a protracted follow-up will be necessary to determine the long-term effects of the treatment on COVID-19 patients.
Collapse
Affiliation(s)
- Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Lucknow, Uttar Pradesh, India.
| | | |
Collapse
|
18
|
Therapeutic Mesenchymal Stromal Cells for Immunotherapy and for Gene and Drug Delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:204-224. [PMID: 32071924 PMCID: PMC7012781 DOI: 10.1016/j.omtm.2020.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) possess several fairly unique properties that, when combined, make them ideally suited for cellular-based immunotherapy and as vehicles for gene and drug delivery for a wide range of diseases and disorders. Key among these are: (1) their relative ease of isolation from a variety of tissues; (2) the ability to be expanded in culture without a loss of functionality, a property that varies to some degree with tissue source; (3) they are relatively immune-inert, perhaps obviating the need for precise donor/recipient matching; (4) they possess potent immunomodulatory functions that can be tailored by so-called licensing in vitro and in vivo; (5) the efficiency with which they can be modified with viral-based vectors; and (6) their almost uncanny ability to selectively home to damaged tissues, tumors, and metastases following systemic administration. In this review, we summarize the latest research in the immunological properties of MSCs, their use as immunomodulatory/anti-inflammatory agents, methods for licensing MSCs to customize their immunological profile, and their use as vehicles for transferring both therapeutic genes in genetic disease and drugs and genes designed to destroy tumor cells.
Collapse
|
19
|
Bone Marrow-Derived Mesenchymal Stromal Cells: A Novel Target to Optimize Hematopoietic Stem Cell Transplantation Protocols in Hematological Malignancies and Rare Genetic Disorders. J Clin Med 2019; 9:jcm9010002. [PMID: 31861268 PMCID: PMC7019991 DOI: 10.3390/jcm9010002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
: Mesenchymal stromal cells (MSCs) are crucial elements in the bone marrow (BM) niche where they provide physical support and secrete soluble factors to control and maintain hematopoietic stem progenitor cells (HSPCs). Given their role in the BM niche and HSPC support, MSCs have been employed in the clinical setting to expand ex-vivo HSPCs, as well as to facilitate HSPC engraftment in vivo. Specific alterations in the mesenchymal compartment have been described in hematological malignancies, as well as in rare genetic disorders, diseases that are amenable to allogeneic hematopoietic stem cell transplantation (HSCT), and ex-vivo HSPC-gene therapy (HSC-GT). Dissecting the in vivo function of human MSCs and studying their biological and functional properties in these diseases is a critical requirement to optimize transplantation outcomes. In this review, the role of MSCs in the orchestration of the BM niche will be revised, and alterations in the mesenchymal compartment in specific disorders will be discussed, focusing on the need to correct and restore a proper microenvironment to ameliorate transplantation procedures, and more in general disease outcomes.
Collapse
|
20
|
Bartold M, Gronthos S, Haynes D, Ivanovski S. Mesenchymal stem cells and biologic factors leading to bone formation. J Clin Periodontol 2019; 46 Suppl 21:12-32. [PMID: 30624807 DOI: 10.1111/jcpe.13053] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/23/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Physiological bone formation and bone regeneration occurring during bone repair can be considered distinct but similar processes. Mesenchymal stem cells (MSC) and associated biologic factors are crucial to both bone formation and bone regeneration. AIM To perform a narrative review of the current literature regarding the role of MSC and biologic factors in bone formation with the aim of discussing the clinical relevance of in vitro and in vivo animal studies. METHODS The literature was searched for studies on MSC and biologic factors associated with the formation of bone in the mandible and maxilla. The search specifically targeted studies on key aspects of how stem cells and biologic factors are important in bone formation and how this might be relevant to bone regeneration. The results are summarized in a narrative review format. RESULTS Different types of MSC and many biologic factors are associated with bone formation in the maxilla and mandible. CONCLUSION Bone formation and regeneration involve very complex and highly regulated cellular and molecular processes. By studying these processes, new clinical opportunities will arise for therapeutic bone regenerative treatments.
Collapse
Affiliation(s)
- Mark Bartold
- School of Dentistry, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - David Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Saso Ivanovski
- School of Dentistry, University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
21
|
Ciciarello M, Corradi G, Loscocco F, Visani G, Monaco F, Cavo M, Curti A, Isidori A. The Yin and Yang of the Bone Marrow Microenvironment: Pros and Cons of Mesenchymal Stromal Cells in Acute Myeloid Leukemia. Front Oncol 2019; 9:1135. [PMID: 31709192 PMCID: PMC6823864 DOI: 10.3389/fonc.2019.01135] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/10/2019] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have, for a long time, been recognized as pivotal contributors in the set up and maintenance of the hematopoietic stem cell (HSC) niche, as well as in the development and differentiation of the lympho-hematopoietic system. MSCs also have a unique immunomodulatory capacity, which makes them able to affect, both in vitro and in vivo, the function of immune cells. These features, namely the facilitation of stem cell engraftment and the inhibition of lymphocyte responses, have both proven essential for successful allogeneic stem cell transplantation (allo-SCT), which remains the only curative option for several hematologic malignancies. For example, in steroid-refractory acute graft-vs. host disease developing after allo-SCT, MSCs have produced significant results and are now considered a treatment option. However, more recently, the other side of the MSC coin has been unveiled, because of their emerging role in creating a protective and immune-tolerant microenvironment able to support the survival of leukemic cells and affect the response to therapies. In this light, it has been proposed that the failure of current treatments to efficiently override the stroma-mediated protection of leukemic cells accounts for the high rate of relapse in acute myeloid leukemia, at least in part. In this review, we will focus on emerging microenvironment-driven mechanisms conferring a survival advantage to leukemic cells overt physiological HSCs. This body of evidence increasingly highlights the opportunity to consider tumor-microenvironment interactions when designing new therapeutic strategies.
Collapse
Affiliation(s)
- Marilena Ciciarello
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Giulia Corradi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Federica Monaco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Hematology and Oncology, Institute of Hematology "L. and A. Seràgnoli", University Hospital S.Orsola-Malpighi, Bologna, Italy
| | - Antonio Curti
- Department of Hematology and Oncology, Institute of Hematology "L. and A. Seràgnoli", University Hospital S.Orsola-Malpighi, Bologna, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| |
Collapse
|
22
|
Chan CKF, Gulati GS, Sinha R, Tompkins JV, Lopez M, Carter AC, Ransom RC, Reinisch A, Wearda T, Murphy M, Brewer RE, Koepke LS, Marecic O, Manjunath A, Seo EY, Leavitt T, Lu WJ, Nguyen A, Conley SD, Salhotra A, Ambrosi TH, Borrelli MR, Siebel T, Chan K, Schallmoser K, Seita J, Sahoo D, Goodnough H, Bishop J, Gardner M, Majeti R, Wan DC, Goodman S, Weissman IL, Chang HY, Longaker MT. Identification of the Human Skeletal Stem Cell. Cell 2019; 175:43-56.e21. [PMID: 30241615 DOI: 10.1016/j.cell.2018.07.029] [Citation(s) in RCA: 401] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 01/16/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Charles K F Chan
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA.
| | - Gunsagar S Gulati
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | | | - Michael Lopez
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Ava C Carter
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Ryan C Ransom
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Andreas Reinisch
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Taylor Wearda
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Matthew Murphy
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Rachel E Brewer
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Lauren S Koepke
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Owen Marecic
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Anoop Manjunath
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Eun Young Seo
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Tripp Leavitt
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Allison Nguyen
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Stephanie D Conley
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Ankit Salhotra
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Thomas H Ambrosi
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Mimi R Borrelli
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Taylor Siebel
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Karen Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Katharina Schallmoser
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Jun Seita
- Medical Sciences Innovation Hub Program, RIKEN, Tokyo 103-0027, Japan
| | - Debashis Sahoo
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Henry Goodnough
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Julius Bishop
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Michael Gardner
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Stuart Goodman
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
Wang W, Han ZC. Heterogeneity of Human Mesenchymal Stromal/Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1123:165-177. [DOI: 10.1007/978-3-030-11096-3_10] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
24
|
Schertl P, Volk J, Perduns R, Adam K, Leyhausen G, Bakopoulou A, Geurtsen W. Impaired angiogenic differentiation of dental pulp stem cells during exposure to the resinous monomer triethylene glycol dimethacrylate. Dent Mater 2018; 35:144-155. [PMID: 30502225 DOI: 10.1016/j.dental.2018.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Dental pulp stem cells (DPSCs) can differentiate into tissue specific lineages to support dental pulp regeneration after injuries. Triethylene glycol dimethacrylate (TEGDMA) is a widely used co-monomer in restorative dentistry with adverse effects on cellular metabolism. Aim of this study was to analyze the impact of TEGDMA on the angiogenic differentiation potential of DPSCs. METHODS DPSCs were characterized by flow cytometry. Short-term (max. 72h) cytotoxicity of TEGDMA was assessed by MTT assay. To evaluate TEGDMA effects on angiogenic differentiation, DPSCs were cultivated in angiogenic differentiation medium (ADM) in the presence or absence of short-term non-toxic TEGDMA concentrations (0.1mM and 0.25mM). Subsequently, angiogenic differentiation was analyzed by qRT-PCR analysis of mRNA markers and in vitro spheroid sprouting assays. RESULTS DPSCs treated with 0.25mM TEGDMA revealed downregulation of angiogenesis-related marker genes PECAM1 (max. 3.8-fold), VEGF-A (max. 2.4-fold) and FLT1 (max. 2.9-fold) compared to respective untreated control. In addition, a reduction of the sprouting potential of DPSCs cultured in the presence of 0.25mM TEGDMA was detectable. Larger spheroidal structures were detectable in the untreated control in comparison to cells treated with 0.25mM TEGDMA. In contrast, TEGDMA at 0.1mM was not affecting angiogenic potential in the investigated time period (up to 28 days). SIGNIFICANCE The results of the present study show that TEGDMA concentration dependently impair the angiogenic differentiation potential of DPSCs and may affect wound healing and the formation of granulation tissue.
Collapse
Affiliation(s)
- Peter Schertl
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, D-30625 Hannover, Germany.
| | - Joachim Volk
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, D-30625 Hannover, Germany.
| | - Renke Perduns
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, D-30625 Hannover, Germany.
| | - Knut Adam
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, D-30625 Hannover, Germany.
| | - Gabriele Leyhausen
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, D-30625 Hannover, Germany.
| | - Athina Bakopoulou
- Department of Fixed Prosthesis and Implant Prosthodontics, School of Dentistry, Aristotle University of Thessaloniki (A.U.TH.), Thessaloniki, Greece.
| | - Werner Geurtsen
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, D-30625 Hannover, Germany.
| |
Collapse
|
25
|
Ranga Rao S, Subbarayan R. Passage-dependent expression of STRO-1 in human gingival mesenchymal stem cells. J Cell Biochem 2018; 120:2810-2815. [PMID: 30260000 DOI: 10.1002/jcb.27674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/21/2018] [Indexed: 11/08/2022]
Abstract
The expression of STRO-1, the essential mesenchymal stem cell marker, was found to decrease with advancing passages in few tissues. Because STRO-1 was identified and isolated from human gingiva, we were interested to know its status after a few passages. Human gingival mesenchymal stem cells (HGMSCs) were isolated from human gingiva. Flow cytometry was carried out with STRO-1, mesenchymal stem cell (MSC) positive marker CD73, and negative marker CD34/CD45. Samples were also subjected to CD90 and STRO-1 immunofluorescence staining. Gene expression was carried out for transcription factors OCT-4, NANOG, and NESTIN. The results showed a gradual decrease in STRO-1 and transcription factor expression with an increase in passage numbers. MSC positive marker CD73 was consistently expressed in all the passages. Negative markers were absent in all the passages. We conclude that STRO-1 may be a useful marker to isolate undifferentiated (potent) mesenchymal cells from gingiva.
Collapse
Affiliation(s)
- Suresh Ranga Rao
- Department of Periodontology and Implantology, Faculty of Dental Sciences & Centre for Regenerative Medicine and Stem Cell Research, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Rajasekaran Subbarayan
- Centre for Regenerative Medicine and Stem Cell Research, Central Research Facility, Sri Ramachandra Medical College and Research Institute, Chennai, India
| |
Collapse
|
26
|
Alfaifi M, Eom YW, Newsome PN, Baik SK. Mesenchymal stromal cell therapy for liver diseases. J Hepatol 2018; 68:1272-1285. [PMID: 29425678 DOI: 10.1016/j.jhep.2018.01.030] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/16/2018] [Accepted: 01/30/2018] [Indexed: 02/08/2023]
Abstract
The therapeutic potential of mesenchymal stromal cells (MSCs) in the treatment of liver fibrosis is predominantly based on their immunosuppressive properties, and their ability to secrete various trophic factors. This potential has been investigated in clinical and preclinical studies. Although the therapeutic mechanisms of MSC transplantation are still not fully characterised, accumulating evidence has revealed that various trophic factors secreted by MSCs play key therapeutic roles in regeneration by alleviating inflammation, apoptosis, and fibrosis as well as stimulating angiogenesis and tissue regeneration in damaged liver. In this review, we summarise the safety, efficacy, potential transplantation routes and therapeutic effects of MSCs in patients with liver fibrosis. We also discuss some of the key strategies to enhance the functionality of MSCs, which include sorting and/or priming with factors such as cytokines, as well as genetic engineering.
Collapse
Affiliation(s)
- Mohammed Alfaifi
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, South Korea; Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Philip N Newsome
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, UK; National Institute for Health Research Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, UK; Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - Soon Koo Baik
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, South Korea; Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea.
| |
Collapse
|
27
|
Ercal P, Pekozer GG, Gumru OZ, Kose GT, Ramazanoglu M. Influence of STRO-1 selection on osteogenic potential of human tooth germ derived mesenchymal stem cells. Arch Oral Biol 2017; 82:293-301. [DOI: 10.1016/j.archoralbio.2017.06.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 01/03/2023]
|
28
|
Janel A, Berger J, Bourgne C, Lemal R, Boiret-Dupré N, Dubois-Galopin F, Déchelotte P, Bothorel C, Hermet E, Chabi S, Bay JO, Lambert C, Pereira B, Pflumio F, Haddad R, Berger MG. Bone marrow hematons: An access point to the human hematopoietic niche. Am J Hematol 2017. [PMID: 28639326 DOI: 10.1002/ajh.24830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To understand the complex interactions between hematopoietic stem cells and the bone marrow niche, a human experimental model is needed. Our hypothesis is that hematons are an appropriate ex vivo model of human bone marrow. We analyzed the hierarchical hematopoietic cell content and the tissue organization of single hematons from healthy donors. Most (>90%) hematons contained precursors of all cell lineages, myeloid progenitors, and LTC-ICs without preferential commitment. Approximately, half of the hematons could generate significant levels of lympho-myeloid hematopoiesis after transplantation in an NSG mouse model, despite the low absolute numbers of transplanted CD34+ cells. Mesenchymal STRO-1+ and/or CD271+ cells formed a critical network that preserved hematon cohesion, and STRO-1+ cells colocalized with most hematopoietic CD34+ cells (68%). We observed an influence of age and gender. These structures represent a particularly attractive model for studying the homeostasis of the bone marrow niche and pathological changes that occur during diseases.
Collapse
Affiliation(s)
- Alexandre Janel
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Biologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
- Université Clermont Auvergne, Equipe d'accueil l'EA 7453 CHELTER; 1 place L. et R. Aubrac, Clermont-Ferrand Cedex 63003 France
| | - Juliette Berger
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Biologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
- Université Clermont Auvergne, Equipe d'accueil l'EA 7453 CHELTER; 1 place L. et R. Aubrac, Clermont-Ferrand Cedex 63003 France
- CHU Clermont-Ferrand, Hôpital Estaing, CRB-Auvergne; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Céline Bourgne
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Biologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
- Université Clermont Auvergne, Equipe d'accueil l'EA 7453 CHELTER; 1 place L. et R. Aubrac, Clermont-Ferrand Cedex 63003 France
| | - Richard Lemal
- Université Clermont Auvergne, Equipe d'accueil l'EA 7453 CHELTER; 1 place L. et R. Aubrac, Clermont-Ferrand Cedex 63003 France
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Clinique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Nathalie Boiret-Dupré
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Biologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Frédérique Dubois-Galopin
- CHU de Toulouse, Hôpital Purpan, Laboratoire d'Hématologie, Place du Docteur Baylac - TSA 40031 31059; Toulouse Cedex 9 France
| | - Pierre Déchelotte
- CHU Clermont-Ferrand, Hôpital Estaing, Anatomie Pathologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Charlotte Bothorel
- CHU Clermont-Ferrand, Hôpital Estaing, Anatomie Pathologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Eric Hermet
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Clinique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Sara Chabi
- INSERM UMR967, CEA/DSV/iRCM, Laboratory of Hematopoietic Stem cells and Leukemic Cells, Equipe labellisée par la Ligue Nationale Contre le Cancer, Université Paris Diderot, Université Paris-Saclay, Univ Paris Sud, Commissariat à l'Energie Atomique et aux Energies Alternatives; Fontenay-aux-Roses 92265 France
| | - Jacques-Olivier Bay
- Université Clermont Auvergne, Equipe d'accueil l'EA 7453 CHELTER; 1 place L. et R. Aubrac, Clermont-Ferrand Cedex 63003 France
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Clinique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Céline Lambert
- CHU Clermont-Ferrand, Département de Recherche Clinique et Innovation, Bd Léon Malfreyt; Clermont-Ferrand France
| | - Bruno Pereira
- CHU Clermont-Ferrand, Département de Recherche Clinique et Innovation, Bd Léon Malfreyt; Clermont-Ferrand France
| | - Françoise Pflumio
- CHU Clermont-Ferrand, Hôpital Estaing, Anatomie Pathologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Rima Haddad
- CHU Clermont-Ferrand, Hôpital Estaing, Anatomie Pathologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| | - Marc G. Berger
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Biologique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
- Université Clermont Auvergne, Equipe d'accueil l'EA 7453 CHELTER; 1 place L. et R. Aubrac, Clermont-Ferrand Cedex 63003 France
- CHU Clermont-Ferrand, Hôpital Estaing, CRB-Auvergne; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
- CHU Clermont-Ferrand, Hôpital Estaing, Hématologie Clinique; 1 place Lucie et Raymond Aubrac, Clermont-Ferrand Cedex 1 63003 France
| |
Collapse
|
29
|
Cytokine treatment optimises the immunotherapeutic effects of umbilical cord-derived MSC for treatment of inflammatory liver disease. Stem Cell Res Ther 2017; 8:140. [PMID: 28595619 PMCID: PMC5465593 DOI: 10.1186/s13287-017-0590-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) possess immunomodulatory properties and low immunogenicity, both crucial properties for their development into an effective cellular immunotherapy. They have shown benefit in clinical trials targeting liver diseases; however the efficacy of MSC therapy will benefit from improvement of the immunomodulatory and immunogenic properties of MSC. METHODS MSC derived from human umbilical cords (ucMSC) were treated for 3 days in vitro with various inflammatory factors, interleukins, vitamins and serum deprivation. Their immunogenicity and immunomodulatory capacity were examined by gene-expression analysis, surface-marker expressions, IDO activity, PGE2 secretion and inhibition of T cell proliferation and IFNγ production. Furthermore, their activation of NK cell cytotoxicity was investigated via CD107a expression on NK cells. The immunomodulatory capacity, biodistribution and survival of pre-treated ucMSC were investigated in a CCl4-induced liver disease mouse model. In addition, capacity of pre-treated MSC to ameliorate liver inflammation was examined in an ex vivo liver inflammation co-culture model. RESULTS IFN-γ and a multiple cytokine cocktail (MC) consisting of IFN-γ, TGFβ and retinoic acid upregulated the expression of immunomodulatory factor PD-L1 and IDO activity. Subsequently, both treatments enhanced the capacity of ucMSC to inhibit CD4 and CD8 T cell proliferation and IFN-γ production. The susceptibility of ucMSC for NK cell lysis was decreased by IFN-β, TGFβ and MC treatment. In vivo, no immunomodulation was observed by the ucMSC. Four hours after intravenous infusion in mice with CCl4-induced inflammatory liver injury, the majority of ucMSC were trapped in the lungs. Rapid clearance of ucMSC(VitB6), ucMSC(Starv + VitB6) and ucMSC(MC) and altered bio-distribution of ucMSC(TGFβ) compared to untreated ucMSC was observed. In the ex vivo co-culture system with inflammatory liver slices ucMSC(MC) showed significantly enhanced modulatory capacity compared to untreated ucMSC. CONCLUSIONS The present study demonstrates the responsiveness of ucMSC to in vitro optimisation treatment. The observed improvements in immunomodulatory capacity as well as immunogenicity after MC treatment may improve the efficacy of ucMSC as immunotherapy targeted towards liver inflammation.
Collapse
|
30
|
Wang F, Jia Y, Liu J, Zhai J, Cao N, Yue W, He H, Pei X. Dental pulp stem cells promote regeneration of damaged neuron cells on the cellular model of Alzheimer's disease. Cell Biol Int 2017; 41:639-650. [PMID: 28328017 DOI: 10.1002/cbin.10767] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/19/2017] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease and many types of stem cells have been used in AD therapy with some favorable effects. In this study, we investigated the potential therapeutical effects of human dental pulp stem cells (hDPSCs) on AD cellular model which established by okadaic acid (OA)-induced damage to human neuroblastoma cell line, SH-SY5Y, in vitro for 24 h. After confirmed the AD cellular model, the cells were co-culture with hDPSCs by transwell co-culture system till 24 h for treatment. Then the cytomorphology of the hDPSCs-treated cells were found to restore gradually with re-elongation of retracted dendrites. Meanwhile, Cell Counting Kit-8 assay and Hoechst 33258 staining showed that hDPSCs caused significant increase in the viability and decrease in apoptosis of the model cells, respectively. Observation of DiI labeling also exhibited the prolongation dendrites in hDPSCs-treated cells which were obviously different from the retraction dendrites in AD model cells. Furthermore, specific staining of α-tubulin and F-actin demonstrated that the hDPSCs-treated cells had the morphology of restored neurons, with elongated dendrites, densely arranged microfilaments, and thickened microtubular fibrils. In addition, results from western blotting revealed that phosphorylation at Ser 396 of Tau protein was significantly suppressed by adding of hDPSCs. These results indicate that hDPSCs may promote regeneration of damaged neuron cells in vitro model of AD and may serve as a useful cell source for treatment of AD.
Collapse
Affiliation(s)
- Feixiang Wang
- Department of Stomatology, Chinese PLA General Hospital and Chinese PLA Medical School, 28, Fuxing Road, Beijing, 100853, China
| | - Yali Jia
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Academy of Military Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Jiajing Liu
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Academy of Military Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Jinglei Zhai
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Academy of Military Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Ning Cao
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Academy of Military Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Academy of Military Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| | - Huixia He
- Department of Stomatology, Chinese PLA General Hospital and Chinese PLA Medical School, 28, Fuxing Road, Beijing, 100853, China
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Academy of Military Medical Sciences, 27, Taiping Road, Beijing, 100850, China
| |
Collapse
|
31
|
Fitter S, Gronthos S, Ooi SS, Zannettino AC. The Mesenchymal Precursor Cell Marker Antibody STRO-1 Binds to Cell Surface Heat Shock Cognate 70. Stem Cells 2017; 35:940-951. [DOI: 10.1002/stem.2560] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Stephen Fitter
- Myeloma Research Laboratory, Faculty of Health and Medical Science, Adelaide Medical School
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide; Adelaide South Australia Australia
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| | - Soo Siang Ooi
- Myeloma Research Laboratory, Faculty of Health and Medical Science, Adelaide Medical School
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| | - Andrew C.W. Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Science, Adelaide Medical School
- Cancer Theme, South Australian Health and Medical Research Institute; Adelaide South Australia Australia
| |
Collapse
|
32
|
Metheny L, Eid S, Lingas K, Reese J, Meyerson H, Tong A, de Lima M, Huang AY. Intra-osseous Co-transplantation of CD34-selected Umbilical Cord Blood and Mesenchymal Stromal Cells. ACTA ACUST UNITED AC 2016; 1:25-29. [PMID: 27882356 PMCID: PMC5117423 DOI: 10.15761/hmo.1000105] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human mesenchymal stromal cells (MSC) have been shown to support the growth and differentiation of hematopoietic stem cells (HSC). We hypothesized that intra-osseous (IO) co-transplantation of MSC and umbilical cord blood (UCB) may be effective in improving early HSC engraftment, as IO transplantation has been demonstrated to enhance UCB engraftment in NOD SCID-gamma (NSG) mice. Following non-lethal irradiation (300rads), 6 groups of NSG mice were studied: 1) intravenous (IV) UCB CD34+ cells, 2) IV UCB CD34+ cells and MSC, 3) IO UCB CD34+ cells, 4) IO UCB CD34+ cells and IO MSC, 5) IO UCB CD34+ cells and IV MSC, and 6) IV UCB CD34+ and IO MSC. Analysis of human-derived CD45+, CD3+, and CD19+ cells 6 weeks following transplant revealed the highest level of engraftment in the IO UCB plus IO MSC cohort. Bone marrow analysis of human CD13 and CD14 markers revealed no significant difference between cohorts. We observed that IO MSC and UCB co-transplantation led to superior engraftment of CD45+, CD3+ and CD19+ lineage cells in the bone marrow at 6 weeks as compared with the IV UCB cohort controls. Our data suggests that IO co-transplantation of MSC and UCB facilitates human HSC engraftment in NSG mice.
Collapse
Affiliation(s)
- Leland Metheny
- Stem Cell Transplant Program, University Hospitals Case Medical Center and Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Saada Eid
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Case Western Reserve University School of Medicine; Angie Fowler AYA Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Karen Lingas
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jane Reese
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Howard Meyerson
- Department of Pathology, Case Western Reserve University School of Medicine; University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Alexander Tong
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Marcos de Lima
- Stem Cell Transplant Program, University Hospitals Case Medical Center and Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Alex Y Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Case Western Reserve University School of Medicine; Angie Fowler AYA Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University School of Medicine; University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
33
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
34
|
Mo M, Wang S, Zhou Y, Li H, Wu Y. Mesenchymal stem cell subpopulations: phenotype, property and therapeutic potential. Cell Mol Life Sci 2016; 73:3311-21. [PMID: 27141940 PMCID: PMC11108490 DOI: 10.1007/s00018-016-2229-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/16/2016] [Accepted: 04/14/2016] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSC) are capable of differentiating into cells of multiple cell lineages and have potent paracrine effects. Due to their easy preparation and low immunogenicity, MSC have emerged as an extremely promising therapeutic agent in regenerative medicine for diverse diseases. However, MSC are heterogeneous with respect to phenotype and function in current isolation and cultivation regimes, which often lead to incomparable experimental results. In addition, there may be specific stem cell subpopulations with definite differentiation capacity toward certain lineages in addition to stem cells with multi-differentiation potential. Recent studies have identified several subsets of MSC which exhibit distinct features and biological activities, and enhanced therapeutic potentials for certain diseases. In this review, we give an overview of these subsets for their phenotypic, biological and functional properties.
Collapse
Affiliation(s)
- Miaohua Mo
- School of Life Sciences, Tsinghua University, Beijing, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, L406A, Tsinghua Campus, The University Town, Shenzhen, China
| | - Shan Wang
- School of Life Sciences, Tsinghua University, Beijing, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, L406A, Tsinghua Campus, The University Town, Shenzhen, China
| | - Ying Zhou
- School of Life Sciences, Tsinghua University, Beijing, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, L406A, Tsinghua Campus, The University Town, Shenzhen, China
| | - Hong Li
- Department of General Surgery, Qingdao Municipal Hospital, 5 Donghai M Rd, Qingdao, China.
| | - Yaojiong Wu
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, L406A, Tsinghua Campus, The University Town, Shenzhen, China.
| |
Collapse
|
35
|
Mishra SK, Rana P, Khushu S, Gangenahalli G. Therapeutic Prospective of Infused Allogenic Cultured Mesenchymal Stem Cells in Traumatic Brain Injury Mice: A Longitudinal Proton Magnetic Resonance Spectroscopy Assessment. Stem Cells Transl Med 2016; 6:316-329. [PMID: 28170180 PMCID: PMC5442758 DOI: 10.5966/sctm.2016-0087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022] Open
Abstract
Improved therapeutic assessment of experimental traumatic brain injury (TBI), using mesenchymal stem cells (MSCs), would immensely benefit its therapeutic management. Neurometabolite patterns at injury site, measured with proton magnetic resonance spectroscopy (1H‐MRS) after MSCs transplantation, may serve as a bio‐indicator of the recovery mechanism. This study used in vivo magnetic resonance imaging and 1H‐MRS to evaluate the therapeutic prospects of implanted MSCs at injury site in experimental mice longitudinally up to 21 days. Negative tissue contrast and cytotoxic edema formation were observed in susceptibility‐based contrast (T2*) and an apparent diffusion coefficient map, respectively. Lesion site showed decreased N‐acetylaspartate, total choline, myo‐inositol, total creatine, glutamate‐glutamine complex, and taurine neurometabolic concentrations by 1H‐MRS investigation. There was a considerable decrease in locomotor activity, depression index, and cognitive index after TBI. It may, therefore, be inferred that MSC transplantation prompted recovery by decreasing negative signals and edema, restoring metabolites to baseline concentrations, and enhancing behavioral activity. Overall findings support the potential of MSC transplantation for the enhancement of endogenous neuroprotective responses, which may provide future clinical applications for translating laboratory research into therapeutic clinical advances. Stem Cells Translational Medicine2017;6:316–329
Collapse
Affiliation(s)
- Sushanta Kumar Mishra
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Defense Research and Development Organisation, Timarpur, Delhi, India
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Timarpur, Delhi, India
| | - Poonam Rana
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Defense Research and Development Organisation, Timarpur, Delhi, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Defense Research and Development Organisation, Timarpur, Delhi, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Timarpur, Delhi, India
| |
Collapse
|
36
|
Futrega K, Lott WB, Doran MR. Direct bone marrow HSC transplantation enhances local engraftment at the expense of systemic engraftment in NSG mice. Sci Rep 2016; 6:23886. [PMID: 27065210 PMCID: PMC4827391 DOI: 10.1038/srep23886] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 03/15/2016] [Indexed: 12/11/2022] Open
Abstract
Direct bone marrow (BM) injection has been proposed as a strategy to bypass homing inefficiencies associated with intravenous (IV) hematopoietic stem cell (HSC) transplantation. Despite physical delivery into the BM cavity, many donor cells are rapidly redistributed by vascular perfusion, perhaps compromising efficacy. Anchoring donor cells to 3-dimensional (3D) multicellular spheroids, formed from mesenchymal stem/stromal cells (MSC) might improve direct BM transplantation. To test this hypothesis, relevant combinations of human umbilical cord blood-derived CD34(+) cells and BM-derived MSC were transplanted into NOD/SCID gamma (NSG) mice using either IV or intrafemoral (IF) routes. IF transplantation resulted in higher human CD45(+) and CD34(+) cell engraftment within injected femurs relative to distal femurs regardless of cell combination, but did not improve overall CD45(+) engraftment at 8 weeks. Analysis within individual mice revealed that despite engraftment reaching near saturation within the injected femur, engraftment at distal hematopoietic sites including peripheral blood, spleen and non-injected femur, could be poor. Our data suggest that the retention of human HSC within the BM following direct BM injection enhances local chimerism at the expense of systemic chimerism in this xenogeneic model.
Collapse
Affiliation(s)
- Kathryn Futrega
- Queensland University of Technology (QUT) at the Translational Research Institute (TRI), 37 Kent Street, Brisbane, Queensland, Australia 4102
| | - William B Lott
- Queensland University of Technology (QUT) at the Translational Research Institute (TRI), 37 Kent Street, Brisbane, Queensland, Australia 4102
| | - Michael R Doran
- Queensland University of Technology (QUT) at the Translational Research Institute (TRI), 37 Kent Street, Brisbane, Queensland, Australia 4102.,Mater Medical Research - University of Queensland at the Translational Research Institute (TRI), 37 Kent Street, Brisbane, Queensland, Australia 4102
| |
Collapse
|
37
|
De Becker A, Riet IV. Homing and migration of mesenchymal stromal cells: How to improve the efficacy of cell therapy? World J Stem Cells 2016; 8:73-87. [PMID: 27022438 PMCID: PMC4807311 DOI: 10.4252/wjsc.v8.i3.73] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/24/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are currently being investigated for use in a wide variety of clinical applications. For most of these applications, systemic delivery of the cells is preferred. However, this requires the homing and migration of MSCs to a target tissue. Although MSC homing has been described, this process does not appear to be highly efficacious because only a few cells reach the target tissue and remain there after systemic administration. This has been ascribed to low expression levels of homing molecules, the loss of expression of such molecules during expansion, and the heterogeneity of MSCs in cultures and MSC culture protocols. To overcome these limitations, different methods to improve the homing capacity of MSCs have been examined. Here, we review the current understanding of MSC homing, with a particular focus on homing to bone marrow. In addition, we summarize the strategies that have been developed to improve this process. A better understanding of MSC biology, MSC migration and homing mechanisms will allow us to prepare MSCs with optimal homing capacities. The efficacy of therapeutic applications is dependent on efficient delivery of the cells and can, therefore, only benefit from better insights into the homing mechanisms.
Collapse
|
38
|
de Witte SFH, Franquesa M, Baan CC, Hoogduijn MJ. Toward Development of iMesenchymal Stem Cells for Immunomodulatory Therapy. Front Immunol 2016; 6:648. [PMID: 26779185 PMCID: PMC4701910 DOI: 10.3389/fimmu.2015.00648] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSC) are under development as an immunomodulatory therapy. The anticipated immunomodulatory effects of MSC are broad, from direct inhibition of lymphocyte proliferation, induction of regulatory T and B cells, to resetting the immune system via a hit-and-run principle. There are endless flavors of MSC. Differences between MSC are originating from donors variation, differences in tissue of origin, the effects of culture conditions, and expansion time. Even standard culture conditions change the properties of MSC dramatically and generate MSC that only remotely resemble their in vivo counterparts. Adjustments in culture protocols can further emphasize properties of interest in MSC, thereby generating cells fitted for specific purposes. Culture improved immunomodulatory MSC can be designed to target particular immune disorders. In this review, we describe the observed and the desired immunomodulatory effects of MSC and propose approaches how MSC with optimal immunomodulatory properties can be developed.
Collapse
Affiliation(s)
- Samantha F H de Witte
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Marcella Franquesa
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Carla C Baan
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| |
Collapse
|
39
|
Yuan YH, Zhou CF, Lu ZY, Wang XL, Ding Y, Li D. Intrabone marrow injection enhances placental mesenchymal stem cellmediated support of hematopoiesis in mice. Turk J Med Sci 2016; 46:174-84. [PMID: 27511352 DOI: 10.3906/sag-1411-161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 04/03/2015] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND/AIM In order to determine the synergistic effects of human placental mesenchymal stem cells (PMSCs) on hematopoiesis in vivo, we compared the intrabone marrow injection (IBMI) with the conventional intravenous injection (IVI). MATERIALS AND METHODS C57BL/6 recipient mice conditioned with lethal doses of irradiation were transplanted with bone marrow mononuclear cells (MNCs) and bone marrow-derived mesenchymal stem cells (BMSCs) from BALB/c mice by IBMI or IVI. NOD/SCID recipient mice conditioned with sublethal doses of irradiation were transplanted with human umbilical cord blood MNCs (UCB-MNCs) and PMSCs by IBMI or IVI. RESULTS The number of hematopoietic cells was significantly higher in mice transplanted with BMSCs by IBMI than in those transplanted by IVI in a murine transplantation model (BALB/c→C57BL/6). Moreover, the percentage of human hematopoietic cells in the tibiae of the NOD/SCID mice that were transplanted with PMSCs plus UCB-MNCs was higher than that in mice transplanted with UCB-MNCs alone. In addition, in mice that were transplanted with PMSCs, PMSCs injected by IBMI were more efficient than those injected by IVI. CONCLUSION Our results not only elucidated the role of PMSCs in promoting hematopoiesis, but also revealed the therapeutic potential of the combination of PMSCs and IBMI in transplantation.
Collapse
Affiliation(s)
- Ya Hong Yuan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Chun Fang Zhou
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Zhi Yong Lu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Xiao Li Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Dongsheng Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, P.R. China
| |
Collapse
|
40
|
Stenger EO, Krishnamurti L, Galipeau J. Mesenchymal stromal cells to modulate immune reconstitution early post-hematopoietic cell transplantation. BMC Immunol 2015; 16:74. [PMID: 26674007 PMCID: PMC4681052 DOI: 10.1186/s12865-015-0135-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent progenitor cells known to modulate the immune system and to promote hematopoiesis. These dual effects make MSCs attractive for use as cellular therapy in hematopoietic cell transplantation (HCT). MSCs can be used peri-HCT or pre-engraftment to modulate immune reconstitution, promoting hematopoietic stem cell (HSC) engraftment and/or preventing graft-versus-host disease (GVHD). Pre-clinical studies have demonstrated that MSCs can potentiate HSC engraftment and prevent GVHD in a variety of animal models. Clinical trials have been small and largely non-randomized but have established safety and early evidence of efficacy, supporting the need for larger randomized trials.
Collapse
Affiliation(s)
- Elizabeth O Stenger
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, 1405 Clifton Road, Atlanta, GA, 30322, USA.
| | - Lakshmanan Krishnamurti
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, 1405 Clifton Road, Atlanta, GA, 30322, USA.
| | - Jacques Galipeau
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, 1405 Clifton Road, Atlanta, GA, 30322, USA. .,Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA.
| |
Collapse
|
41
|
Zheng YL, Sun YP, Zhang H, Liu WJ, Jiang R, Li WY, Zheng YH, Zhang ZG. Mesenchymal Stem Cells Obtained from Synovial Fluid Mesenchymal Stem Cell-Derived Induced Pluripotent Stem Cells on a Matrigel Coating Exhibited Enhanced Proliferation and Differentiation Potential. PLoS One 2015; 10:e0144226. [PMID: 26649753 PMCID: PMC4674106 DOI: 10.1371/journal.pone.0144226] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 11/16/2015] [Indexed: 01/27/2023] Open
Abstract
Induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) serve as a promising source for cell-based therapies in regenerative medicine. However, optimal methods for transforming iPSCs into MSCs and the characteristics of iPSC-MSCs obtained from different methods remain poorly understood. In this study, we developed a one-step method for obtaining iPSC-MSCs (CD146+STRO-1+ MSCs) from human synovial fluid MSC-derived induced iPSCs (SFMSC-iPSCs). CD146-STRO-1-SFMSCs were reprogrammed into iPSCs by transduction with lentivirus-mediated Sox2, Oct-3/4, klf4, and c-Myc. SFMSC-iPSCs were maintained with mTeSR1 medium in Matrigel-coated culture plates. Single dissociated cells were obtained by digesting the SFMSC-iPSCs with trypsin. The dissociated cells were then plated into Matrigel-coated culture plate with alpha minimum essential medium supplemented with 10% fetal bovine serum, 1× Glutamax, and the ROCK inhibitor Y-27632. Cells were then passaged in standard cell culture plates with alpha minimum essential medium supplemented with 10% fetal bovine serum and 1× Glutamax. After passaging in vitro, the cells showed a homogenous spindle-shape similar to their ancestor cells (SFMSCs), but with more robust proliferative activity. Flow cytometric analysis revealed typical MSC surface markers, including expression of CD73, CD90, CD105, and CD44 and lack of CD45, CD34, CD11b, CD19, and HLA-DR. However, these cells were positive for CD146 and stro-1, which the ancestor cells were not. Moreover, the cells could also be induced to differentiate in osteogenic, chondrogenic, and adipogenic lineages in vitro. The differentiation potential was improved compared with the ancestor cells in vitro. The cells were not found to exhibit oncogenicity in vivo. Therefore, the method presented herein facilitated the generation of STRO-1+CD146+ MSCs from SFMSC-iPSCs exhibiting enhanced proliferation and differentiation potential.
Collapse
Affiliation(s)
- Yu-Liang Zheng
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
- Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou, Guangdong, P.R. China
| | - Yang-Peng Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
- * E-mail: (ZZ); (YS)
| | - Hong Zhang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Wen-Jing Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Rui Jiang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Wen-Yu Li
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - You-Hua Zheng
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Zhi-Guang Zhang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
- * E-mail: (ZZ); (YS)
| |
Collapse
|
42
|
García-García A, de Castillejo CLF, Méndez-Ferrer S. BMSCs and hematopoiesis. Immunol Lett 2015; 168:129-35. [PMID: 26192443 DOI: 10.1016/j.imlet.2015.06.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 01/05/2023]
Abstract
Recent discoveries have significantly expanded our previous knowledge about the role of bone marrow mesenchymal stem cells (BMSCs) in hematopoiesis. BMSCs and their derivatives modulate blood production and immunity at different levels but a prominent role has emerged for BMSCs in the regulation of hematopoietic stem and progenitor cells (HSPCs). Additionally, BMSC-like cells regulate B and T cell lymphopoiesis and also probably myelopoiesis. Furthermore, BMSCs might also exhibit key regulatory properties in non-physiological conditions. BMSCs in extramedullary sites might provide a permissive microenvironment to allow for transient hematopoiesis. BMSCs might be also involved in the manifestation and/or the development of hematopoietic diseases, as stemming from their emerging roles in the progression of hematological malignancies. Here we review some key molecular pathways, adhesion molecules and ligand/receptor interactions that mediate the crosstalk between BMSCs and hematopoietic stem cells (HSCs) in health and disease. The development of novel markers to visualize and isolate individual cells will help to dissect the stromal-hematopoietic interplay.
Collapse
Affiliation(s)
- Andrés García-García
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; Stem Cell Institute and Department of Haematology, University of Cambridge, and National Health Service Blood and Transplant, Cambridge Biomedical Campus, CB20PT Cambridge, United Kingdom
| | - Carlos L F de Castillejo
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; Stem Cell Institute and Department of Haematology, University of Cambridge, and National Health Service Blood and Transplant, Cambridge Biomedical Campus, CB20PT Cambridge, United Kingdom
| | - Simón Méndez-Ferrer
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; Stem Cell Institute and Department of Haematology, University of Cambridge, and National Health Service Blood and Transplant, Cambridge Biomedical Campus, CB20PT Cambridge, United Kingdom.
| |
Collapse
|
43
|
Organ-specific migration of mesenchymal stromal cells: Who, when, where and why? Immunol Lett 2015; 168:159-69. [DOI: 10.1016/j.imlet.2015.06.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/17/2015] [Accepted: 06/23/2015] [Indexed: 12/13/2022]
|
44
|
Reichert D, Friedrichs J, Ritter S, Käubler T, Werner C, Bornhäuser M, Corbeil D. Phenotypic, Morphological and Adhesive Differences of Human Hematopoietic Progenitor Cells Cultured on Murine versus Human Mesenchymal Stromal Cells. Sci Rep 2015; 5:15680. [PMID: 26498381 PMCID: PMC4620509 DOI: 10.1038/srep15680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023] Open
Abstract
Xenogenic transplantation models have been developed to study human hematopoiesis in immunocompromised murine recipients. They still have limitations and therefore it is important to delineate all players within the bone marrow that could account for species-specific differences. Here, we evaluated the proliferative capacity, morphological and physical characteristics of human CD34+ hematopoietic stem and progenitor cells (HSPCs) after co-culture on murine or human bone marrow-derived mesenchymal stromal cells (MSCs). After seven days, human CD34+CD133– HSPCs expanded to similar extents on both feeder layers while cellular subsets comprising primitive CD34+CD133+ and CD133+CD34– phenotypes are reduced fivefold on murine MSCs. The number of migrating HSPCs was also reduced on murine cells suggesting that MSC adhesion influences cellular polarization of HSPC. We used atomic force microscopy-based single-cell force spectroscopy to quantify their adhesive interactions. We found threefold higher detachment forces of human HSPCs from murine MSCs compared to human ones. This difference is related to the N-cadherin expression level on murine MSCs since its knockdown abolished their differential adhesion properties with human HSPCs. Our observations highlight phenotypic, morphological and adhesive differences of human HSPCs when cultured on murine or human MSCs, which raise some caution in data interpretation when xenogenic transplantation models are used.
Collapse
Affiliation(s)
- Doreen Reichert
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Jens Friedrichs
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Steffi Ritter
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Theresa Käubler
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Carsten Werner
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, 01307 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| | - Denis Corbeil
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| |
Collapse
|
45
|
Jones E, Schäfer R. Where is the common ground between bone marrow mesenchymal stem/stromal cells from different donors and species? Stem Cell Res Ther 2015; 6:143. [PMID: 26282627 PMCID: PMC4539918 DOI: 10.1186/s13287-015-0144-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) feature promising potential for cellular therapies, yet significant progress in development of MSC therapeutics and assays is hampered because of remarkable MSC heterogeneity in vivo and in vitro. This heterogeneity poses challenges for standardization of MSC characterization and potency assays as well as for MSC study comparability and manufacturing. This review discusses promising marker combinations for prospective MSC subpopulation enrichment and expansion, and reflects MSC phenotype changes due to environment and age. In order to address animal modelling in MSC biology, comparison of mouse and human MSC markers highlights current common ground of MSCs between species.
Collapse
Affiliation(s)
- Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds University, Room 5.24 Clinical Sciences Building, St James's University Hospital, Leeds, LS9 7TF, UK.
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service, Baden-Württemberg-Hessen gGmbH, Johann-Wolfgang-Goethe University Hospital, Sandhofstrasse 1, D-60528, Frankfurt am Main, Germany.
| |
Collapse
|
46
|
Wu CG, Zhang JC, Xie CQ, Parolini O, Silini A, Huang YZ, Lian B, Zhang M, Huang YC, Deng L. In vivo tracking of human placenta derived mesenchymal stem cells in nude mice via ¹⁴C-TdR labeling. BMC Biotechnol 2015; 15:55. [PMID: 26070459 PMCID: PMC4465458 DOI: 10.1186/s12896-015-0174-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 05/29/2015] [Indexed: 02/05/2023] Open
Abstract
Background In order to shed light on the regenerative mechanism of mesenchymal stem cells (MSCs) in vivo, the bio-distribution profile of implanted cells using a stable and long-term tracking method is needed. We herein investigated the bio-distribution of human placental deciduas basalis derived MSCs (termed as PDB-MSCs) in nude mice after intravenous injection by carbon radioisotope labeling thymidine (14C-TdR), which is able to incorporate into new DNA strands during cell replication. Results The proliferation rate and radioactive emission of human PDB-MSCs after labeled with different concentrations of 14C-TdR were measured. PDB-MSCs labeled with 1 μCi possessed high radioactivity, and the biological characteristics (i.e. morphology, colony forming ability, differentiation capabilities, karyotype and cell cycle) showed no significant changes after labeling. Thus, 1 μCi was the optimal concentration in this experimental design. In nude mice, 1 × 10614C-TdR-labeled PDB-MSCs were injected intravenously and the organs were collected at days 1, 2, 3, 5, 30 and 180 after injection, respectively. Radiolabeled PDB-MSCs were found mainly in the lung, liver, spleen, stomach and left femur of the recipient nude mice at the whole observation period. Conclusions This work provided solid evidence that 14C-TdR labeling did not alter the biological characteristics of human placental MSCs, and that this labeling method has potential to decrease the signal from non-infused or dead cells for cell tracking. Therefore, this labeling technique can be utilized to quantify the infused cells after long-term follow-up in pre-clinical studies.
Collapse
Affiliation(s)
- Cheng-Guang Wu
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Ji-Chun Zhang
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Cheng-Quan Xie
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Ornella Parolini
- Centro di Ricerca E.Menni, Fondazione Poliambulanza, Brescia, Italy.
| | - Antonietta Silini
- Centro di Ricerca E.Menni, Fondazione Poliambulanza, Brescia, Italy.
| | - Yi-Zhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Bing Lian
- West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China.
| | - Min Zhang
- Center Laboratory For Isotopy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Yong-Can Huang
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| | - Li Deng
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
47
|
Modulating Mesenchymal Stem Cell Behavior Using Human Hair Keratin-Coated Surfaces. Stem Cells Int 2015; 2015:752424. [PMID: 26124842 PMCID: PMC4466490 DOI: 10.1155/2015/752424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 01/29/2015] [Accepted: 03/29/2015] [Indexed: 11/18/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) have shown great potential for therapeutic purposes. However, the low frequencies of hMSCs in the body and difficulties in expanding their numbers in vitro have limited their clinical use. In order to develop an alternative strategy for the expansion of hMSCs in vitro, we coated tissue culture polystyrene with keratins extracted from human hair and studied the behavior of cells from 2 donors on these surfaces. The coating resulted in a homogeneous distribution of nanosized keratin globules possessing significant hydrophilicity. Results from cell attachment assays demonstrated that keratin-coated surfaces were able to moderate donor-to-donor variability when compared with noncoated tissue culture polystyrene. STRO-1 expression was either sustained or enhanced on hMSCs cultured on keratin-coated surfaces. This translated into significant increases in the colony-forming efficiencies of both hMSC populations, when the cells were serially passaged. Human hair keratins are abundant and might constitute a feasible replacement for other biomaterials that are of animal origin. In addition, our results suggest that hair keratins may be effective in moderating the microenvironment sufficiently to enrich hMSCs with high colony-forming efficiency ex vivo, for clinical applications.
Collapse
|
48
|
Ejtehadifar M, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Dehdilani N, Abbasi P, Molaeipour Z, Saleh M. The Effect of Hypoxia on Mesenchymal Stem Cell Biology. Adv Pharm Bull 2015; 5:141-9. [PMID: 26236651 DOI: 10.15171/apb.2015.021] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 09/12/2014] [Accepted: 09/17/2014] [Indexed: 12/27/2022] Open
Abstract
Although physiological and pathological role of hypoxia have been appreciated in mammalians for decades however the cellular biology of hypoxia more clarified in the past 20 years. Discovery of the transcription factor hypoxia-inducible factor (HIF)-1, in the 1990s opened a new window to investigate the mechanisms behind hypoxia. In different cellular contexts HIF-1 activation show variable results by impacting various aspects of cell biology such as cell cycle, apoptosis, differentiation and etc. Mesenchymal stem cells (MSC) are unique cells which take important role in tissue regeneration. They are characterized by self-renewal capacity, multilineage potential, and immunosuppressive property. Like so many kind of cells, hypoxia induces different responses in MSCs by HIF- 1 activation. The activation of this molecule changes the growth, multiplication, differentiation and gene expression profile of MSCs in their niche by a complex of signals. This article briefly discusses the most important effects of hypoxia in growth kinetics, signalling pathways, cytokine secretion profile and expression of chemokine receptors in different conditions.
Collapse
Affiliation(s)
- Mostafa Ejtehadifar
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. ; Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tabriz, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Drug Applied Research Center and Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Dehdilani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvaneh Abbasi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Molaeipour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahshid Saleh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
New Steps in the Use of Mesenchymal Stem Cell in Solid Organ Transplantation. CURRENT TRANSPLANTATION REPORTS 2015. [DOI: 10.1007/s40472-015-0053-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
50
|
Lv FJ, Tuan RS, Cheung KMC, Leung VYL. Concise review: the surface markers and identity of human mesenchymal stem cells. Stem Cells 2015; 32:1408-19. [PMID: 24578244 DOI: 10.1002/stem.1681] [Citation(s) in RCA: 730] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 02/09/2014] [Indexed: 12/13/2022]
Abstract
The concept of mesenchymal stem cells (MSCs) is becoming increasingly obscure due to the recent findings of heterogeneous populations with different levels of stemness within MSCs isolated by traditional plastic adherence. MSCs were originally identified in bone marrow and later detected in many other tissues. Currently, no cloning based on single surface marker is capable of isolating cells that satisfy the minimal criteria of MSCs from various tissue environments. Markers that associate with the stemness of MSCs await to be elucidated. A number of candidate MSC surface markers or markers possibly related to their stemness have been brought forward so far, including Stro-1, SSEA-4, CD271, and CD146, yet there is a large difference in their expression in various sources of MSCs. The exact identity of MSCs in vivo is not yet clear, although reports have suggested they may have a fibroblastic or pericytic origin. In this review, we revisit the reported expression of surface molecules in MSCs from various sources, aiming to assess their potential as MSC markers and define the critical panel for future investigation. We also discuss the relationship of MSCs to fibroblasts and pericytes in an attempt to shed light on their identity in vivo.
Collapse
Affiliation(s)
- Feng-Juan Lv
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China; Stem Cell & Regenerative Medicine Consortium, The University of Hong Kong, Hong Kong SAR, People's Republic of China; Center for Reproduction, Development and Growth, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | |
Collapse
|