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
|
Smolinska A, Chodkowska M, Kominek A, Janiec J, Piwocka K, Sulejczak D, Sarnowska A. Stemness properties of SSEA-4+ subpopulation isolated from heterogenous Wharton's jelly mesenchymal stem/stromal cells. Front Cell Dev Biol 2024; 12:1227034. [PMID: 38455073 PMCID: PMC10917976 DOI: 10.3389/fcell.2024.1227034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 01/17/2024] [Indexed: 03/09/2024] Open
Abstract
Background: High heterogeneity of mesenchymal stem/stromal cells (MSCs) due to different degrees of differentiation of cell subpopulations poses a considerable challenge in preclinical studies. The cells at a pluripotent-like stage represent a stem cell population of interest for many researchers worldwide, which is worthy of identification, isolation, and functional characterization. In the current study, we asked whether Wharton's jelly-derived MSCs (WJ-MSCs) which express stage-specific embryonic antigen-4 (SSEA-4) can be considered as a pluripotent-like stem cell population. Methods: SSEA-4 expression in different culture conditions was compared and the efficiency of two cell separation methods were assessed: Magnetic Activated Cell Sorting (MACS) and Fluorescence Activated Cell Sorting (FACS). After isolation, SSEA-4+ cells were analyzed for the following parameters: the maintenance of the SSEA-4 antigen expression after cell sorting, stem cell-related gene expression, proliferation potential, clonogenicity, secretome profiling, and the ability to form spheres under 3D culture conditions. Results: FACS allowed for the enrichment of SSEA-4+ cell content in the population that lasted for six passages after sorting. Despite the elevated expression of stemness-related genes, SSEA-4+ cells neither differed in their proliferation and clonogenicity potential from initial and negative populations nor exhibited pluripotent differentiation repertoire. SSEA-4+ cells were observed to form smaller spheroids and exhibited increased survival under 3D conditions. Conclusion: Despite the transient expression of stemness-related genes, our findings could not fully confirm the undifferentiated pluripotent-like nature of the SSEA-4+ WJ-MSC population cultured in vitro.
Collapse
Affiliation(s)
- Agnieszka Smolinska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Chodkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Agata Kominek
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jakub Janiec
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
3
|
Zong Q, Bundkirchen K, Neunaber C, Noack S. Effect of High BMI on Human Bone Marrow-Derived Mesenchymal Stromal Cells. Cell Transplant 2024; 33:9636897241226546. [PMID: 38258516 PMCID: PMC10807335 DOI: 10.1177/09636897241226546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) are attractive candidates in tissue engineering and regenerative medicine. Growing evidence has suggested that a high body mass index (BMI) can affect the properties of BMSCs, resulting in a reduced quality of the cells. However, the results are not consistent. Therefore, this study aimed to investigate the influences of high BMI on human BMSCs (hBMSCs). To avoid gender bias, BMSCs from females and males were studied independently. Finally, hBMSCs from 89 females and 152 males were separately divided into the normal BMI group (18.5 kg/m2 ≤ BMI < 25 kg/m2) and the high BMI group (BMI > 25 kg/m2). The cells were analyzed for the colony-forming potential; proliferation capacity; in vitro adipogenic, osteogenic, and chondrogenic differentiation potentials; and the expression of 32 common surface antigens. The results showed that high BMI did not change the number of colonies at passage 1 in females and males. In contrast, significantly reduced colony numbers at passage 4 (P4) were found in both female and male donors with high BMI. The doubling time of hBMSCs was comparable between the normal and the high BMI groups of females and males. Furthermore, the results of trilineage differentiation did not differ between the different BMI groups of males. In females, the high and the normal BMI groups also showed similar adipogenic and chondrogenic differentiation, while osteogenic differentiation was significantly enhanced in the high-BMI group. Regarding the expression of surface antigens, the expressions of CD200 and SSEA4 on hBMSCs were reduced in the high-BMI group of females and males, respectively. In conclusion, high BMI suppressed the clonogenicity of female and male hBMSCs at P4, improved the in vitro osteogenesis of female hBMSCs, and decreased the expressions of CD200 on hBMSCs in females and SSEA4 in males.
Collapse
Affiliation(s)
- Qiang Zong
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Katrin Bundkirchen
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Claudia Neunaber
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Sandra Noack
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Smolinska A, Bzinkowska A, Rybkowska P, Chodkowska M, Sarnowska A. Promising Markers in the Context of Mesenchymal Stem/Stromal Cells Subpopulations with Unique Properties. Stem Cells Int 2023; 2023:1842958. [PMID: 37771549 PMCID: PMC10533301 DOI: 10.1155/2023/1842958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/11/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023] Open
Abstract
The heterogeneity of the mesenchymal stem/stromal cells (MSCs) population poses a challenge to researchers and clinicians, especially those observed at the population level. What is more, the lack of precise evidences regarding MSCs developmental origin even further complicate this issue. As the available evidences indicate several possible pathways of MSCs formation, this diverse origin may be reflected in the unique subsets of cells found within the MSCs population. Such populations differ in specialization degree, proliferation, and immunomodulatory properties or exhibit other additional properties such as increased angiogenesis capacity. In this review article, we attempted to identify such outstanding populations according to the specific surface antigens or intracellular markers. Described groups were characterized depending on their specialization and potential therapeutic application. The reports presented here cover a wide variety of properties found in the recent literature, which is quite scarce for many candidates mentioned in this article. Even though the collected information would allow for better targeting of specific subpopulations in regenerative medicine to increase the effectiveness of MSC-based therapies.
Collapse
Affiliation(s)
- Agnieszka Smolinska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Aleksandra Bzinkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Paulina Rybkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Magdalena Chodkowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| |
Collapse
|
5
|
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
|
6
|
Selle M, Koch JD, Ongsiek A, Ulbrich L, Ye W, Jiang Z, Krettek C, Neunaber C, Noack S. Influence of age on stem cells depends on the sex of the bone marrow donor. J Cell Mol Med 2022; 26:1594-1605. [PMID: 35088539 PMCID: PMC8899192 DOI: 10.1111/jcmm.17201] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/05/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022] Open
Abstract
Ageing is often accompanied by an increase in bone marrow fat together with reduced bone volume and diseases of the bone such as osteoporosis. As mesenchymal stem cells (MSCs) are capable of forming bone, cartilage and fat tissue, studying these cells is of great importance to understand the underlying mechanisms behind age‐related bone diseases. However, inter‐donor variation has been found when handling MSCs. Therefore, the aim of this study was to investigate the effects of donor age and sex by comparing in vitro characteristics of human bone marrow‐derived MSCs (hBMSCs) from a large donor cohort (n = 175). For this, hBMSCs were analysed for CFU‐F capacity, proliferation, differentiation capacity and surface antigen expression under standardized culture conditions. The results demonstrated a significantly reduced CFU‐F number for hBMSCs of female compared to male donors. Furthermore, there was a significant decrease in the proliferation rate, adipogenic differentiation potential and cell surface expression of SSEA‐4, CD146 and CD274 of hBMSCs with an increase in donor age. Interestingly, all these findings were exclusive to hBMSCs from female donors. Further research should focus on postmenopausal‐related effects on hBMSCs, as the results imply a functional loss and immunophenotypic change of hBMSCs particularly in aged women.
Collapse
Affiliation(s)
- Michael Selle
- Trauma Department, Hannover Medical School, Hannover, Germany
| | | | - Alina Ongsiek
- Trauma Department, Hannover Medical School, Hannover, Germany
| | - Linnea Ulbrich
- Trauma Department, Hannover Medical School, Hannover, Germany
| | - Weikang Ye
- Trauma Department, Hannover Medical School, Hannover, Germany
| | - Zhida Jiang
- Trauma Department, Hannover Medical School, Hannover, Germany
| | | | | | - Sandra Noack
- Trauma Department, Hannover Medical School, Hannover, Germany
| |
Collapse
|
7
|
Wang T, Zhang J, Liao J, Zhang F, Zhou G. Donor genetic backgrounds contribute to the functional heterogeneity of stem cells and clinical outcomes. Stem Cells Transl Med 2020; 9:1495-1499. [PMID: 32830917 PMCID: PMC7695629 DOI: 10.1002/sctm.20-0155] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/21/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022] Open
Abstract
Stable and sustainable stem cell sources for stem cell‐based therapies are scarce and a key bottleneck for clinical applications. The regenerative potential of stem cells is usually attributed to several allogeneic or even autologous donor‐related factors. Genetic background and epigenetic variations in different individuals may significantly affect the functional heterogeneity of stem cells. Particularly, single‐nucleotide polymorphisms (SNPs) have been implicated in diseases with monogenetic or multifactorial and complex genetic etiologies. However, the possible effects of individual SNPs on donor stem cells remain far from fully elucidated. In this Perspective, we will discuss the roles played by donor genetic traits in the functional heterogeneity of induced pluripotent stem cells, mesenchymal stem cells, and hematopoietic stem cells and their implications for regenerative medicine and therapy.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, People's Republic of China
| | - Juan Zhang
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, People's Republic of China.,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Jinqi Liao
- Lungene Scientific Ltd., Shenzhen, People's Republic of China
| | - Fan Zhang
- Department of Endocrinology and Metabolic Diseases, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, People's Republic of China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, People's Republic of China
| |
Collapse
|
8
|
Yin L, Yang Z, Wu Y, Denslin V, Yu CC, Tee CA, Lim CT, Han J, Lee EH. Label-free separation of mesenchymal stem cell subpopulations with distinct differentiation potencies and paracrine effects. Biomaterials 2020; 240:119881. [PMID: 32092592 DOI: 10.1016/j.biomaterials.2020.119881] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells (MSCs) have the capability to differentiate into multiple cell lineages, and produce trophic factors to facilitate tissue repair and regeneration, and disease regression. However, the heterogeneity of MSCs, whether inherent or developed during culture expansion, has a significant impact on their therapeutic efficacy. Therefore, the ability to identify and select an efficacious subpopulation of MSCs targeting specific tissue damage or disease holds great clinical significance. In this study, we separated three subpopulations from culture expanded human bone marrow derived MSCs according to cell size, using a high-throughput label-free microfluidic cell sorting technology. The size-sorted MSC subpopulations varied in tri-lineage differentiation potencies. The large MSCs showed the strongest osteogenesis, medium-size MSCs were advantageous in chondrogenesis and adipogenesis, and the small MSCs showed the weakest tri-lineage differentiation. The size-sorted MSC subpopulations also exhibited different secretome profiles. The large MSC secretome possessed highest levels of osteogenic promotor proteins and senescence-associated factors, but lower levels of osteogenic inhibitor proteins compared to the medium-size MSC secretome. The medium-size MSC secretome had high levels of chondrogenic promotor proteins, and contained lower levels of chondrogenic inhibitor proteins compared to the large MSC secretome. The secretome of size-sorted MSC subpopulations showed differences in paracrine effects. We found that the secretome of large MSCs enhanced osteogenic and adipogenic potencies during MSC culture expansion, but also induced cell senescence; and the secretome of medium-size MSCs promoted chondrogenesis. This study demonstrates size-dependent differentiation potency and secretome profile of MSC subpopulations, and provides an effective and practical technology to isolate the respective subpopulations, which may be used for more targeted tissue repair and regeneration.
Collapse
Affiliation(s)
- Lu Yin
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore
| | - Zheng Yang
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Yingnan Wu
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Vinitha Denslin
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Chia Chen Yu
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Ching Ann Tee
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, T-Lab, #10-01, Singapore, 117411, Singapore; Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore; Institute for Health Innovation and Technology, National University of Singapore, MD6, 14 Medical Drive, #14-01, Singapore, 117599, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Department of Electrical Engineering and Computer Science, Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Eng Hin Lee
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore.
| |
Collapse
|
9
|
Dalton S, Smith K, Singh K, Kaiser H, Kolhe R, Mondal AK, Khayrullin A, Isales CM, Hamrick MW, Hill WD, Fulzele S. Accumulation of kynurenine elevates oxidative stress and alters microRNA profile in human bone marrow stromal cells. Exp Gerontol 2020; 130:110800. [PMID: 31790802 PMCID: PMC6998036 DOI: 10.1016/j.exger.2019.110800] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 02/09/2023]
Abstract
Kynurenine, a metabolite of tryptophan breakdown, has been shown to increase with age, and plays a vital role in a number of age-related pathophysiological changes, including bone loss. Accumulation of kynurenine in bone marrow stromal cells (BMSCs) has been associated with a decrease in cell proliferation and differentiation, though the exact mechanism by which kynurenine mediates these changes is poorly understood. MiRNAs have been shown to regulate BMSC function, and accumulation of kynurenine may alter the miRNA expression profile of BMSCs. The aim of this study was to identify differentially expressed miRNAs in human BMSCs in response to treatment with kynurenine, and correlate miRNAs function in BMSCs biology through bioinformatics analysis. Human BMSCs were cultured and treated with and without kynurenine, and subsequent miRNA isolation was performed. MiRNA array was performed to identify differentially expressed miRNA. Microarray analysis identified 50 up-regulated, and 36 down-regulated miRNAs in kynurenine-treated BMSC cultures. Differentially expressed miRNA included miR-1281, miR-330-3p, let-7f-5p, and miR-493-5p, which are important for BMSC proliferation and differentiation. KEGG analysis found up-regulated miRNA targeting glutathione metabolism, a pathway critical for removing oxidative species. Our data support that the kynurenine dependent degenerative effect is partially due to changes in the miRNA profile of BMSCs.
Collapse
Affiliation(s)
- Sherwood Dalton
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Kathryn Smith
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Kanwar Singh
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America
| | - Helen Kaiser
- Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Ravindra Kolhe
- Departments of Pathology, Augusta University, Augusta, GA 30912, United States of America
| | - Ashis K Mondal
- Departments of Pathology, Augusta University, Augusta, GA 30912, United States of America
| | - Andrew Khayrullin
- Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Medicine, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC, 29403, United States of America
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States of America; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States of America; Institute of Healthy Aging, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
10
|
O'Connor KC. Molecular Profiles of Cell-to-Cell Variation in the Regenerative Potential of Mesenchymal Stromal Cells. Stem Cells Int 2019; 2019:5924878. [PMID: 31636675 PMCID: PMC6766122 DOI: 10.1155/2019/5924878] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/20/2019] [Indexed: 12/22/2022] Open
Abstract
Cell-to-cell variation in the regenerative potential of mesenchymal stromal cells (MSCs) impedes the translation of MSC therapies into clinical practice. Cellular heterogeneity is ubiquitous across MSC cultures from different species and tissues. This review highlights advances to elucidate molecular profiles that identify cell subsets with specific regenerative properties in heterogeneous MSC cultures. Cell surface markers and global signatures are presented for proliferation and differentiation potential, as well as immunomodulation and trophic properties. Key knowledge gaps are discussed as potential areas of future research. Molecular profiles of MSC heterogeneity have the potential to enable unprecedented control over the regenerative potential of MSC therapies through the discovery of new molecular targets and as quality attributes to develop robust and reproducible biomanufacturing processes. These advances would have a positive impact on the nascent field of MSC therapeutics by accelerating the development of therapies with more consistent and effective treatment outcomes.
Collapse
Affiliation(s)
- Kim C. O'Connor
- Department of Chemical and Biomolecular Engineering, Tulane University, New Orleans, Louisiana, USA
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
11
|
Molecular characterization of mesenchymal stem cells in human osteoarthritis cartilage reveals contribution to the OA phenotype. Sci Rep 2018; 8:7044. [PMID: 29728632 PMCID: PMC5935742 DOI: 10.1038/s41598-018-25395-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 04/16/2018] [Indexed: 11/10/2022] Open
Abstract
Adult human articular cartilage harbors a population of CD166+ mesenchymal stem cell-like progenitors that become more numerous during osteoarthritis (OA). While their role is not well understood, here we report that they are indeed part of cellular clusters formed in OA cartilage, which is a pathological hallmark of this disease. We hypothesize that these cells, termed OA mesenchymal stem cells (OA-MSCs), contribute to OA pathogenesis. To test this hypothesis, we generated and characterized multiple clonally derived stable/immortalized human OA-MSC cell lines, which exhibited the following properties. Firstly, two mesenchymal stem cell populations exist in human OA cartilage. While both populations are multi-potent, one preferentially undergoes chondrogenesis while the other exhibits higher osteogenesis potential. Secondly, both OA-MSCs exhibit significantly higher expression of hypertrophic OA cartilage markers COL10A1 and RUNX2, compared to OA chondrocytes. Induction of chondrogenesis in OA-MSCs further stimulated COL10A1 expression and MMP-13 release, suggesting that they contribute to OA phenotypes. Finally, knocking down RUNX2 is insufficient to inhibit COL10A1 in OA-MSCs and also requires simultaneous knockdown of NOTCH1 thereby suggesting altered gene regulation in OA stem cells in comparison to chondrocytes. Overall, our findings suggest that OA-MSCs may drive pathogenesis of cartilage degeneration and should therefore be a novel cell target for OA therapy.
Collapse
|
12
|
Weist R, Flörkemeier T, Roger Y, Franke A, Schwanke K, Zweigerdt R, Martin U, Willbold E, Hoffmann A. Differential Expression of Cholinergic System Components in Human Induced Pluripotent Stem Cells, Bone Marrow-Derived Multipotent Stromal Cells, and Induced Pluripotent Stem Cell-Derived Multipotent Stromal Cells. Stem Cells Dev 2018; 27:166-183. [PMID: 29205106 DOI: 10.1089/scd.2017.0162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The components of the cholinergic system are evolutionary very old and conserved molecules that are expressed in typical spatiotemporal patterns. They are involved in signaling in the nervous system, whereas their functions in nonneuronal tissues are hardly understood. Stem cells present an attractive cellular system to address functional issues. This study therefore compared human induced pluripotent stem cells (iPSCs; from cord blood endothelial cells), mesenchymal stromal cells derived from iPSCs (iPSC-MSCs), and bone marrow-derived MSCs (BM-MSCs) from up to 33 different human donors with respect to gene expressions of components of the cholinergic system. The status of cells was identified and characterized by the detection of cell surface antigens using flow cytometry. Acetylcholinesterase expression in iPSCs declined during their differentiation into MSCs and was comparably low in BM-MSCs. Butyrylcholinesterase was present in iPSCs, increased upon transition from the three-dimensional embryoid body phase into monolayer culture, and declined upon further differentiation into iPSC-MSCs. In BM-MSCs a notable butyrylcholinesterase expression could be detected in only four donors, but was elusive in other patient-derived samples. Different nicotinic acetylcholine receptor subunits were preferentially expressed in iPSCs and during early differentiation into iPSC-MSCs, low expression was detected in iPS-MSCs and in BM-MSCs. The m2 and m3 variants of muscarinic acetylcholine receptors were detected in all stem cell populations. In BM-MSCs, these gene expressions varied between donors. Together, these data reveal the differential expression of cholinergic signaling system components in stem cells from specific sources and suggest the utility of our approach to establish informative biomarkers.
Collapse
Affiliation(s)
- Ramona Weist
- 1 Department of Orthopaedic Surgery, Graded Implants and Regenerative Strategies, Hannover Medical School , Hannover, Germany .,2 Department of Trauma Surgery, Hannover Medical School , Hannover, Germany
| | - Thilo Flörkemeier
- 3 Laboratory for Biomechanics and Biomaterials, Department of Orthopaedic Surgery, Hannover Medical School , Hannover, Germany
| | - Yvonne Roger
- 1 Department of Orthopaedic Surgery, Graded Implants and Regenerative Strategies, Hannover Medical School , Hannover, Germany .,4 Lower Saxony Centre for Biomedical Engineering , Implant Research and Development (NIFE), Hannover, Germany
| | - Annika Franke
- 5 Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School , Hannover, Germany .,6 REBIRTH-Cluster of Excellence, Hannover Medical School , Hannover, Germany
| | - Kristin Schwanke
- 5 Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School , Hannover, Germany .,6 REBIRTH-Cluster of Excellence, Hannover Medical School , Hannover, Germany
| | - Robert Zweigerdt
- 5 Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School , Hannover, Germany .,6 REBIRTH-Cluster of Excellence, Hannover Medical School , Hannover, Germany
| | - Ulrich Martin
- 5 Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School , Hannover, Germany .,6 REBIRTH-Cluster of Excellence, Hannover Medical School , Hannover, Germany
| | - Elmar Willbold
- 3 Laboratory for Biomechanics and Biomaterials, Department of Orthopaedic Surgery, Hannover Medical School , Hannover, Germany .,4 Lower Saxony Centre for Biomedical Engineering , Implant Research and Development (NIFE), Hannover, Germany
| | - Andrea Hoffmann
- 1 Department of Orthopaedic Surgery, Graded Implants and Regenerative Strategies, Hannover Medical School , Hannover, Germany .,4 Lower Saxony Centre for Biomedical Engineering , Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
13
|
Viter R, Jekabsons K, Kalnina Z, Poletaev N, Hsu SH, Riekstina U. Bioanalytical system for detection of cancer cells with photoluminescent ZnO nanorods. NANOTECHNOLOGY 2016; 27:465101. [PMID: 27731308 DOI: 10.1088/0957-4484/27/46/465101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Using photoluminescent ZnO nanorods and carbohydrate marker SSEA-4, a novel cancer cell recognition system was developed. Immobilization of SSEA-4 antibodies (αSSEA-4) on ZnO nanorods was performed in buffer solution (pH = 7.1) over 2 h. The cancer cell line probes were fixed on the glass slide. One hundred microliters of ZnO-αSSEA-4 conjugates were deposited on the cell probe and exposed for 30 min. After washing photoluminescence spectra were recorded. Based on the developed methodology, ZnO-αSSEA-4 probes were tested on patient-derived breast and colorectal carcinoma cells. Our data clearly show that the carbohydrate SSEA-4 molecule is expressed on cancer cell lines and patient-derived cancer cells. Moreover, SSEA-4 targeted ZnO nanorods bind to the patient-derived cancer cells with high selectivity and the photoluminescence signal increased tremendously compared to the signal from the control samples. Furthermore, the photoluminescence intensity increase correlated with the extent of malignancy in the target cell population. A novel portable bioanalytical system, based on optical ZnO nanorods and fiber optic detection system was developed. We propose that carbohydrate SSEA-4 specific ZnO nanorods could be used for the development of cancer diagnostic biosensors and for targeted therapy.
Collapse
Affiliation(s)
- R Viter
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Riga, Latvia
| | | | | | | | | | | |
Collapse
|
14
|
In Vitro Differentiation of First Trimester Human Umbilical Cord Perivascular Cells into Contracting Cardiomyocyte-Like Cells. Stem Cells Int 2016; 2016:7513252. [PMID: 27123009 PMCID: PMC4829731 DOI: 10.1155/2016/7513252] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/30/2016] [Accepted: 02/11/2016] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction (MI) causes an extensive loss of heart muscle cells and leads to congestive heart disease (CAD), the leading cause of mortality and morbidity worldwide. Mesenchymal stromal cell- (MSC-) based cell therapy is a promising option to replace invasive interventions. However the optimal cell type providing significant cardiac regeneration after MI is yet to be found. The aim of our study was to investigate the cardiomyogenic differentiation potential of first trimester human umbilical cord perivascular cells (FTM HUCPVCs), a novel, young source of immunoprivileged mesenchymal stromal cells. Based on the expression of cardiomyocyte markers (cTnT, MYH6, SIRPA, and CX43) FTM and term HUCPVCs achieved significantly increased cardiomyogenic differentiation compared to bone marrow MSCs, while their immunogenicity remained significantly lower as indicated by HLA-A and HLA-G expression and susceptibility to T cell mediated cytotoxicity. When applying aggregate-based differentiation, FTM HUCPVCs showed increased aggregate formation potential and generated contracting cells within 1 week of coculture, making them the first MSC type with this ability. Our results indicate that young FTM HUCPVCs have superior cardiomyogenic potential coupled with beneficial immunogenic properties when compared to MSCs of older tissue sources, suggesting that in vitro predifferentiation could be a potential strategy to increase their effectiveness in vivo.
Collapse
|
15
|
KARAÇALI S. Human embryonic stem cell N-glycan features relevant to pluripotency. Turk J Biol 2016. [DOI: 10.3906/biy-1509-57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
16
|
Chandran P, Le Y, Li Y, Sabloff M, Mehic J, Rosu-Myles M, Allan DS. Mesenchymal stromal cells from patients with acute myeloid leukemia have altered capacity to expand differentiated hematopoietic progenitors. Leuk Res 2015; 39:486-93. [PMID: 25703353 DOI: 10.1016/j.leukres.2015.01.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 01/01/2023]
Abstract
The bone marrow microenvironment may be permissive to the emergence and progression of acute myeloid leukemia (AML). Studying interactions between the microenvironment and leukemia cells should provide new insight for therapeutic advances. Mesenchymal stromal cells (MSCs) are central to the maintenance of the hematopoietic niche. Here we compared the functions and gene expression patterns of MSCs derived from bone marrow aspirates of healthy donors and patients with AML. MSCs expanded from AML patients had heterogeneous morphology and displayed a wide range of proliferation capacity compared to MSCs from healthy controls. The ability of AML-MSCs to support the expansion of committed hematopoietic progenitors from umbilical cord blood-derived CD34+ cells may be impaired while the expression of genes associated with maintaining hematopoietic quiescence appeared to be increased in AML-MSCs compared to healthy donors. These results highlight important potential differences in the biologic profile of MSCs from AML patients compared to healthy donors that may contribute to the emergence or progression of leukemia.
Collapse
Affiliation(s)
- Priya Chandran
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada
| | - Yevgeniya Le
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada; Atomic Energy of Canada Limited, Chalk River, ON, Canada
| | - Yuhua Li
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada
| | - Mitchell Sabloff
- Department of Medicine, Hematology, University of Ottawa, Ottawa, ON, Canada
| | - Jelica Mehic
- Centre for Biologics Evaluation, Health Canada, Ottawa, Canada
| | | | - David S Allan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada; Department of Medicine, Hematology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
17
|
An B, Na S, Lee S, Kim WJ, Yang SR, Woo HM, Kook S, Hong Y, Song H, Hong SH. Non-enzymatic isolation followed by supplementation of basic fibroblast growth factor improves proliferation, clonogenic capacity and SSEA-4 expression of perivascular cells from human umbilical cord. Cell Tissue Res 2014; 359:767-77. [DOI: 10.1007/s00441-014-2066-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 11/13/2014] [Indexed: 12/20/2022]
|
18
|
Altman E, Yango P, Moustafa R, Smith JF, Klatsky PC, Tran ND. Characterization of human spermatogonial stem cell markers in fetal, pediatric, and adult testicular tissues. Reproduction 2014; 148:417-27. [PMID: 25030892 PMCID: PMC4599365 DOI: 10.1530/rep-14-0123] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autologous spermatogonial stem cell (SSC) transplantation is a potential therapeutic modality for patients with azoospermia following cancer treatment. For this promise to be realized, definitive membrane markers of prepubertal and adult human SSCs must be characterized in order to permit SSC isolation and subsequent expansion. This study further characterizes the markers of male gonocytes, prespermatogonia, and SSCs in humans. Human fetal, prepubertal, and adult testicular tissues were analyzed by confocal microscopy, fluorescence-activated cell sorting, and qRT-PCR for the expression of unique germ cell membrane markers. During male fetal development, THY1 and KIT (C-Kit) are transient markers of gonocytes but not in prespermatogonia and post-natal SSCs. Although KIT expression is detected in gonocytes, THY1 expression is also detected in the somatic component of the fetal testes in addition to gonocytes. In the third trimester of gestation, THY1 expression shifts exclusively to the somatic cells of the testes where it continues to be detected only in the somatic cells postnatally. In contrast, SSEA4 expression was only detected in the gonocytes, prespermatogonia, SSCs, and Sertoli cells of the fetal and prepubertal testes. After puberty, SSEA4 expression can only be detected in primitive spermatogonia. Thus, although THY1 and KIT are transient markers of gonocytes, SSEA4 is the only common membrane marker of gonocytes, prespermatogonia, and SSCs from fetal through adult human development. This finding is essential for the isolation of prepubertal and adult SSCs, which may someday permit fertility preservation and reversal of azoospermia following cancer treatment.
Collapse
Affiliation(s)
- Eran Altman
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Pamela Yango
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Radwa Moustafa
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - James F Smith
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Peter C Klatsky
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Nam D Tran
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| |
Collapse
|
19
|
Smith JF, Yango P, Altman E, Choudhry S, Poelzl A, Zamah AM, Rosen M, Klatsky PC, Tran ND. Testicular niche required for human spermatogonial stem cell expansion. Stem Cells Transl Med 2014; 3:1043-54. [PMID: 25038247 DOI: 10.5966/sctm.2014-0045] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prepubertal boys treated with high-dose chemotherapy do not have an established means of fertility preservation because no established in vitro technique exists to expand and mature purified spermatogonial stem cells (SSCs) to functional sperm in humans. In this study, we define and characterize the unique testicular cellular niche required for SSC expansion using testicular tissues from men with normal spermatogenesis. Highly purified SSCs and testicular somatic cells were isolated by fluorescence-activated cell sorting using SSEA-4 and THY1 as markers of SSCs and somatic cells. Cells were cultured on various established niches to assess their role in SSC expansion in a defined somatic cellular niche. Of all the niches examined, cells in the SSEA-4 population exclusively bound to adult testicular stromal cells, established colonies, and expanded. Further characterization of these testicular stromal cells revealed distinct mesenchymal markers and the ability to undergo differentiation along the mesenchymal lineage, supporting a testicular multipotent stromal cell origin. In vitro human SSC expansion requires a unique niche provided exclusively by testicular multipotent stromal cells with mesenchymal properties. These findings provide an important foundation for developing methods of inducing SSC growth and maturation in prepubertal testicular tissue, essential to enabling fertility preservation for these boys.
Collapse
Affiliation(s)
- James F Smith
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Pamela Yango
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Eran Altman
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Shweta Choudhry
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Andrea Poelzl
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Alberuni M Zamah
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Mitchell Rosen
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Peter C Klatsky
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Nam D Tran
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| |
Collapse
|
20
|
Wu X, Luo Y, Chen J, Pan R, Xiang B, Du X, Xiang L, Shao J, Xiang C. Transplantation of human menstrual blood progenitor cells improves hyperglycemia by promoting endogenous progenitor differentiation in type 1 diabetic mice. Stem Cells Dev 2014; 23:1245-57. [PMID: 24499421 DOI: 10.1089/scd.2013.0390] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recently, a unique population of progenitor cells was isolated from human menstrual blood. The human menstrual blood progenitor cells (MBPCs) possess many advantages, such as the noninvasive acquisition procedure, broad multipotency, a higher proliferative rate, and low immunogenicity, and have attracted extensive attention in regenerative medicine. Preclinical studies to test the safety and efficacy of MBPCs have been underway in several animal models. However, relevant studies in type 1 diabetes mellitus (T1DM) have not yet been proceeded. Herein, we studied the therapeutic effect of MBPCs and the mechanism of β-cell regeneration after MBPC transplantation in the T1DM model. Intravenous injection of MBPCs can reverse hyperglycemia and weight loss, prolong lifespan, and increase insulin production in diabetic mice. Histological and immunohistochemistry analyses indicated that T1DM mice with MBPC transplantation recovered islet structures and increased the β-cell number. We further analyzed in vivo distribution of MBPCs and discovered that a majority of MBPCs migrated into damaged pancreas and located at the islet, duct, and exocrine tissue. MBPCs did not differentiate into insulin-producing cells, but enhanced neurogenin3 (ngn3) expression, which represented endocrine progenitors that were activated. Ngn3(+) cells were not only in the ductal epithelium, but also in the islet and exocrine tissue. We analyzed a series of genes associated with the embryonic mode of β-cell development by real-time polymerase chain reaction and the results showed that the levels of those gene expressions all increased after cell transplantation. According to the results, we concluded that MBPCs stimulated β-cell regeneration through promoting differentiation of endogenous progenitor cells.
Collapse
Affiliation(s)
- Xiaoxing Wu
- 1 State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Can arthroscopically harvested synovial stem cells be preferentially sorted using stage-specific embryonic antigen 4 antibody for cartilage, bone, and adipose regeneration? Arthroscopy 2014; 30:352-61. [PMID: 24581260 DOI: 10.1016/j.arthro.2013.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of this study was to investigate the relation between stage-specific embryonic antigen 4 (SSEA4) expression and synovium-derived stem cell (SDSC) lineage differentiation. METHODS Human SDSCs were collected during arthroscopic surgery from 4 young patients with anterior cruciate ligament injuries. Passage 2 SDSCs were sorted by fluorescence-activated cell sorting using phycoerythrin-conjugated monoclonal antibody against SSEA4 into 3 groups: SSEA4(+) cells, SSEA4(-) cells, and unsorted control cells. After 1 more passage, expanded cells from each group were evaluated for SSEA4 expression by use of flow cytometry as well as multilineage differentiation capacities, including chondrogenesis, adipogenesis, and osteogenesis, using biochemical analysis, histologic analysis, immunostaining, and real-time polymerase chain reaction. RESULTS After cell sorting, 1 more passage expansion decreased SSEA4(+) cells from 99.8% to 79.2% and increased SSEA4(-) cells from 4.4% to 53.3% compared with 70.3% in the unsorted cell population. SSEA4(-) SDSCs with a lower cell proliferation exhibited higher chondrogenic potential (in terms of the ratio of glycosaminoglycan to DNA [P < .001] and COL2A1 [type II collagen] messenger RNA [mRNA] [P < .001]) and adipogenic potential (in terms of oil red O staining and quantitative assay [P = .007], LPL [lipoprotein lipase] mRNA [P = .005], and CEBP [CCAAT/enhancer-binding protein alpha] mRNA [P = .010]). In contrast, SSEA4(+) SDSCs retained cell expansion and enhanced osteogenic capacity, as evidenced by intense calcium deposition stained by alizarin red S and a significantly elevated expression of OPN (osteopontin) mRNA (P = .007). CONCLUSIONS In this study, for the first time, we showed the benefit of using the surface marker SSEA4 in SDSCs to preferentially sort a mixed population of cells. SSEA4(+) SDSCs indicated a strong potential for osteogenesis rather than chondrogenesis and adipogenesis. CLINICAL RELEVANCE SDSC-based mesenchymal tissue regeneration can be easily achieved by arthroscopic harvesting followed by quick cell sorting.
Collapse
|