1
|
Tian L, Wang W, Li X, Chen Y, Song Q, Yuan L, Hao T, Gu J, Dong J. Whole transcriptome scanning and validation of negatively related genes in UC-MSCs. Heliyon 2024; 10:e27996. [PMID: 38510024 PMCID: PMC10951646 DOI: 10.1016/j.heliyon.2024.e27996] [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: 04/21/2023] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
Background Human umbilical cord mesenchymal stem cells (UC-MSCs) are one of the most extensively researched stem cell types due to their potential for multi-lineage differentiation, secretion of regenerative factors, modulations of immunological activities, and the release of regenerative substances and influence immunological processes. Since UC-MSCs must be cultivated on a large scale for clinical use, selecting the appropriate storing passage, such as the usage-based passage of UC-MSCs, is critical for long-term autologous or allogeneic usage. Long-term cultivation of stem cells, on the other hand, causes them to lose their pluripotent differentiation capacity. As a result, distinguishing between high and low passages of UC-MSCs and identifying the particular variations associated with stem cells and their modes of action is essential for regenerative medicine. Therefore, we investigated the biological features and transcriptional changes of UC-MSCs over passages. Methods UC-MSCs were isolated from the tissues of the human umbilical cord, and UC-MSCs from five passages (P1, P3, P5, P10 and P15) with three repetitions were compared and identified based on morphology, cell markers, differentiation capacity, and aging-related characteristics. It was previously assumed that the phenotype of cells before the P10 passage was stable, defined as early passage, and that culture could be continued until the 15th passage, defined as late passage. Next, the five passages of UC-MSCs were sequenced using high-throughput complete transcriptome sequencing. Fuzzy C-Means Clustering (FCM) and Weighted Gene Co-expression Network Analysis (WGCNA) were used to find hub genes, and gene silencing was performed to investigate the impact of missing genes on the stemness of UC-MSC cells. Results UC-MSCs of different passages displayed similar surface markers, including CD73, CD105, CD90, CD34, CD45 and HLA-DR. However, the proliferation time of late-phase UC-MSCs was longer than that of early-phase UC-MSCs, and the expression of the senescence-associated (SA)-β-gal staining marker was higher. At the same time, pluripotency markers (NANOG, OCT4, SOX2 and KIF4A) were down-regulated, and the multi-differentiation potential was reduced. Meanwhile, KIFC1 and UBE2C were down-regulated in late-phase UC-MSCs, which were involved in the maintenance of stemness. Conclusions KIFC1 and UBE2C were highly expressed in early-UC-MSCs and showed a downward gradient trend with cell expansion in vitro. They regulated UC-MSC proliferation, colony sphere formation, multiple differentiation, stemness maintenance, and other biological manifestations. Therefore, they are anticipated to be new biomarkers for UC-MSCs quality identification in regenerative medicine applications.
Collapse
Affiliation(s)
- Linghan Tian
- Department of Yunnan Tumor Research Institute, Kunming, 650118, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
- Yunnan Cancer Hospital, Kunming, 650118, China
| | - Weibin Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Yunnan Research Institute for Local Plateau Agriculture and Industry, Kunming, 650201, China
| | - Xuzhen Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Yunnan Research Institute for Local Plateau Agriculture and Industry, Kunming, 650201, China
| | - Yan Chen
- Department of Yunnan Tumor Research Institute, Kunming, 650118, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
- Yunnan Cancer Hospital, Kunming, 650118, China
| | - Qian Song
- Department of Yunnan Tumor Research Institute, Kunming, 650118, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
- Yunnan Cancer Hospital, Kunming, 650118, China
| | - Lu Yuan
- Department of Yunnan Tumor Research Institute, Kunming, 650118, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
- Yunnan Cancer Hospital, Kunming, 650118, China
| | - Tingting Hao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Yunnan Research Institute for Local Plateau Agriculture and Industry, Kunming, 650201, China
| | - Jiaming Gu
- Department of Yunnan Tumor Research Institute, Kunming, 650118, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
- Yunnan Cancer Hospital, Kunming, 650118, China
| | - Jian Dong
- Department of Yunnan Tumor Research Institute, Kunming, 650118, China
- The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
- Yunnan Cancer Hospital, Kunming, 650118, China
| |
Collapse
|
2
|
He L, Li M, Wang X, Wu X, Yue G, Wang T, Zhou Y, Lei B, Zhou G. Morphology-based deep learning enables accurate detection of senescence in mesenchymal stem cell cultures. BMC Biol 2024; 22:1. [PMID: 38167069 PMCID: PMC10762950 DOI: 10.1186/s12915-023-01780-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Cell senescence is a sign of aging and plays a significant role in the pathogenesis of age-related disorders. For cell therapy, senescence may compromise the quality and efficacy of cells, posing potential safety risks. Mesenchymal stem cells (MSCs) are currently undergoing extensive research for cell therapy, thus necessitating the development of effective methods to evaluate senescence. Senescent MSCs exhibit distinctive morphology that can be used for detection. However, morphological assessment during MSC production is often subjective and uncertain. New tools are required for the reliable evaluation of senescent single cells on a large scale in live imaging of MSCs. RESULTS We have developed a successful morphology-based Cascade region-based convolution neural network (Cascade R-CNN) system for detecting senescent MSCs, which can automatically locate single cells of different sizes and shapes in multicellular images and assess their senescence state. Additionally, we tested the applicability of the Cascade R-CNN system for MSC senescence and examined the correlation between morphological changes with other senescence indicators. CONCLUSIONS This deep learning has been applied for the first time to detect senescent MSCs, showing promising performance in both chronic and acute MSC senescence. The system can be a labor-saving and cost-effective option for screening MSC culture conditions and anti-aging drugs, as well as providing a powerful tool for non-invasive and real-time morphological image analysis integrated into cell production.
Collapse
Affiliation(s)
- Liangge He
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, 1066 Xueyuan Avenue, Shenzhen, 518060, China
- Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopedic Diseases, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Mingzhu Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, 1066 Xueyuan Avenue, Shenzhen, 518060, China
| | - Xinglie Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, 1066 Xueyuan Avenue, Shenzhen, 518060, China
| | - Xiaoyan Wu
- Department of Dermatology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Guanghui Yue
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, 1066 Xueyuan Avenue, Shenzhen, 518060, China
| | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, 1066 Xueyuan Avenue, Shenzhen, 518060, China
| | - Yan Zhou
- Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopedic Diseases, Shenzhen University Medical School, Shenzhen, 518060, China
- Lungene Biotech Ltd., Shenzhen, 18000, China
| | - Baiying Lei
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, 1066 Xueyuan Avenue, Shenzhen, 518060, China.
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopedic Diseases, Shenzhen University Medical School, Shenzhen, 518060, China.
| |
Collapse
|
3
|
Wang J, Qin W, Zhong Y, Hu H, Yang J, Huang H, Huang N, Liu S, Li J, Zheng L, Qin A, Lu Z. Injectable collagen hydrogel combines human umbilical cord mesenchymal stem cells to promote endometrial regeneration in rats with thin endometrium. Int J Biol Macromol 2024; 254:127591. [PMID: 37884246 DOI: 10.1016/j.ijbiomac.2023.127591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/10/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
The regeneration of thin endometrium still remains as a great challenge in the field of reproductive medicine. Stem cells-based therapy has been considered as a promising strategy for the restoration of thin endometrium. However, the low transplantation and retention rate of stem cells and loss of stemness due to in vitro expansion limits the therapeutic efficacy. In our study, we combined collagen hydrogel and human umbilical cord mesenchymal stem cells (uMSCs) for improving the regeneration of thin endometrium, by using the potent pluripotency and low immunogenicity of uMSCs and collagen hydrogel that promotes the anchorage and proliferation of stem cells. Results showed that collagen hydrogel has favorable biocompatibility and the capacity to enhance the cell viability and expression of stemness-associated genes (including organic cation/carnitine transporter4 (Oct-4), Nanog homeobox (Nanog) and SRY-box transcription factor 2 (SOX2)) of uMSCs. The combination of collagen hydrogel and uMSCs prolonged the retention time of the constructs in the uterine cavity and improved endometrial thickness compared with uMSCs alone, leading to increase the fertility of the rats with thin endometrium. These highlighted therapeutic prospects of collagen hydrogel combined with uMSCs for the minimally invasive therapy of thin endometrium in the clinic.
Collapse
Affiliation(s)
- Jiawei Wang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Weili Qin
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yanping Zhong
- Institute of Life Science, Guangxi Medical University, Nanning 530021, China
| | - Hao Hu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Junxu Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hanji Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Institute of Life Science, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Nanchang Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Shuhan Liu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiaxu Li
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Institute of Life Science, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Aiping Qin
- Center of Reproductive Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Institute of Life Science, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, The First Afliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
4
|
Okoro PD, Frayssinet A, De Oliveira S, Rouquier L, Miklosic G, D'Este M, Potier E, Hélary C. Combining biomimetic collagen/hyaluronan hydrogels with discogenic growth factors promotes mesenchymal stroma cell differentiation into Nucleus Pulposus like cells. Biomater Sci 2023; 11:7768-7783. [PMID: 37870786 DOI: 10.1039/d3bm01025b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Based on stem cell injection into degenerated Nucleus Pulposus (NP), novel treatments for intervertebral disc (IVD) regeneration were disappointing because of cell leakage or inappropriate cell differentiation. In this study, we hypothesized that mesenchymal stromal cells encapsulated within injectable hydrogels possessing adequate physico-chemical properties would differentiate into NP like cells. Composite hydrogels consisting of type I collagen and tyramine-substituted hyaluronic acid (THA) were prepared to mimic the NP physico-chemical properties. Human bone marrow derived mesenchymal stromal cells (BM-MSCs) were encapsulated within hydrogels and cultivated in proliferation medium (supplemented with 10% fetal bovine serum) or differentiation medium (supplemented with GDF5 and TGFβ1) over 28 days. Unlike pure collagen, collagen/THA composite hydrogels were stable over 28 days in culture. In proliferation medium, the cell viability within pure collagen hydrogels was high, whereas that in composite and pure THA hydrogels was lower due to the weaker cell adhesion. Nonetheless, BM-MSCs proliferated in all hydrogels. In composite hydrogels, cells exhibited a rounded morphology similar to NP cells. The differentiation medium did not impact the hydrogel stability and cell morphology but negatively impacted the cell viability in pure collagen hydrogels. A high THA content within hydrogels promoted the gene expression of NP markers such as collagen II, aggrecan, SOX9 and cytokeratin 18 at day 28. The differentiation medium potentialized this effect with an earlier and higher expression of these NP markers. Taken together, these results show that the physico-chemical properties of collagen/THA composite hydrogels and GDF5/TGFβ1 act in synergy to promote the differentiation of BM-MSCs into NP like cells.
Collapse
Affiliation(s)
- Prince David Okoro
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France.
| | - Antoine Frayssinet
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France.
| | - Stéphanie De Oliveira
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France.
| | - Léa Rouquier
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, F-75010 Paris, France
| | - Gregor Miklosic
- AO Research Institute Davos (ARI), Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Matteo D'Este
- AO Research Institute Davos (ARI), Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Esther Potier
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, F-75010 Paris, France
| | - Christophe Hélary
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, CNRS, UMR 7574, F-75005, Paris, France.
| |
Collapse
|
5
|
Romantsik O, Moreira A, Thébaud B, Ådén U, Ley D, Bruschettini M. Stem cell-based interventions for the prevention and treatment of intraventricular haemorrhage and encephalopathy of prematurity in preterm infants. Cochrane Database Syst Rev 2023; 2:CD013201. [PMID: 36790019 PMCID: PMC9932000 DOI: 10.1002/14651858.cd013201.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
BACKGROUND Germinal matrix-intraventricular haemorrhage (GMH-IVH) and encephalopathy of prematurity (EoP) remain substantial issues in neonatal intensive care units worldwide. Current therapies to prevent or treat these conditions are limited. Stem cell-based therapies offer a potential therapeutic approach to repair, restore, or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal studies. This is an update of the 2019 review, which did not include EoP. OBJECTIVES To evaluate the benefits and harms of stem cell-based interventions for prevention or treatment of GM-IVH and EoP in preterm infants. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search was April 2022. SELECTION CRITERIA We attempted to include randomised controlled trials, quasi-randomised controlled trials, and cluster trials comparing 1. stem cell-based interventions versus control; 2. mesenchymal stromal cells (MSCs) of type or source versus MSCs of other type or source; 3. stem cell-based interventions other than MSCs of type or source versus stem cell-based interventions other than MSCs of other type or source; or 4. MSCs versus stem cell-based interventions other than MSCs. For prevention studies, we included extremely preterm infants (less than 28 weeks' gestation), 24 hours of age or less, without ultrasound diagnosis of GM-IVH or EoP; for treatment studies, we included preterm infants (less than 37 weeks' gestation), of any postnatal age, with ultrasound diagnosis of GM-IVH or with EoP. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were 1. all-cause neonatal mortality, 2. major neurodevelopmental disability, 3. GM-IVH, 4. EoP, and 5. extension of pre-existing non-severe GM-IVH or EoP. We planned to use GRADE to assess certainty of evidence for each outcome. MAIN RESULTS We identified no studies that met our inclusion criteria. Three studies are currently registered and ongoing. Phase 1 trials are described in the 'Excluded studies' section. AUTHORS' CONCLUSIONS No evidence is currently available to evaluate the benefits and harms of stem cell-based interventions for treatment or prevention of GM-IVH or EoP in preterm infants. We identified three ongoing studies, with a sample size range from 20 to 200. In two studies, autologous cord blood mononuclear cells will be administered to extremely preterm infants via the intravenous route; in one, intracerebroventricular injection of MSCs will be administered to preterm infants up to 34 weeks' gestational age.
Collapse
Affiliation(s)
- Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Ulrika Ådén
- Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
6
|
Xu X, Yin F, Guo M, Gan G, Lin G, Wen C, Wang J, Song P, Wang J, Qi ZQ, Zhong CQ. Quantitative proteomic analysis of exosomes from umbilical cord mesenchymal stem cells and rat bone marrow stem cells. Proteomics 2023; 23:e2200204. [PMID: 36408942 DOI: 10.1002/pmic.202200204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/23/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022]
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) have been used for cancer treatment, however, an in-depth analysis of the exosomal proteomes is lacking. In this manuscript, we use the diaPASEF (parallel accumulation serial fragmentation combined with the data-independent acquisition) method to quantify exosomes derived from human umbilical cord mesenchymal stem cells (UCMSCs) and rat bone marrow stem cells (BMSCs), resulting in identification of 4200 human proteins and 5362 rat proteins. Comparison of human exosomal proteins and total cellular proteins reveals that some proteins exist in the exosomes exclusively that can be served as potential markers for exosomes. Quantitative proteomic analysis of exosomes from different passages of BMSCs shows that the proteins involved in TGF-β signaling pathway are regulated in abundance, which could be markers for the therapeutic ability of BMSC exosomes. Collectively, the data presented by this study can be a resource for further study of exosome research.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Biomedical Engineering, Medical College of Guangxi University, Nanning, Guangxi, China
| | - Fengyue Yin
- Department of Biomedical Engineering, Medical College of Guangxi University, Nanning, Guangxi, China
| | - Mengyu Guo
- Department of Emergency, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
| | - Guohong Gan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Guanzhong Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chengwen Wen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Junsheng Wang
- Department of Emergency, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
| | - Pengbo Song
- Department of Biomedical Engineering, Medical College of Guangxi University, Nanning, Guangxi, China
| | - Jinling Wang
- Department of emergency, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zhong-Quan Qi
- Department of Biomedical Engineering, Medical College of Guangxi University, Nanning, Guangxi, China
| | - Chuan-Qi Zhong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
7
|
Wang S, Wang Z, Su H, Chen F, Ma M, Yu W, Ye G, Cen S, Mi R, Wu X, Deng W, Feng P, Zeng C, Shen H, Wu Y. Effects of long-term culture on the biological characteristics and RNA profiles of human bone-marrow-derived mesenchymal stem cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:557-574. [PMID: 34631285 PMCID: PMC8479280 DOI: 10.1016/j.omtn.2021.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 08/12/2021] [Indexed: 12/16/2022]
Abstract
Expansion in vitro prior to mesenchymal stem cells (MSCs) application is a necessary process. Functional and genomic stability has a crucial role in stem-cell-based therapies. However, the exact expression and co-expressed profiles of coding and non-coding RNAs in human bone marrow (BM)-MSCs in vitro aging are still lacking. In the present studies, the change of morphology, immunophenotype, and capacity of proliferation, differentiation, and immunoregulation of MSCs at passage (P) 4, P6, P8, P10, and P12 were investigated. RNA sequencing identified that 439 mRNAs, 65 long noncoding RNAs (lncRNAs), 59 microRNAs (miRNAs), and 229 circular RNAs (circRNAs) were differentially expressed (DE) in P12 compared with P4, with a similar trend in P6. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) identified several significant biological processes and pathways, including binding, ossification, and Wnt and PPAR signaling pathways. Interaction and co-expression/localization analyses were performed for DE mRNAs and lncRNAs, and several key lncRNAs, circRNAs, and important pathways like autophagy and mitophagy were identified in the competing endogenous RNA (ceRNA) network. Some key RNAs found in the bioinformatics analysis were validated. Our studies indicate that replicative senescence of MSCs is a continuous process, including widespread alterations in biological characteristics and global gene expression patterns that need to be considered before therapeutic applications of MSCs.
Collapse
Affiliation(s)
- Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Hongjun Su
- Center for Biotherapy, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Mengjun Ma
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Wenhui Yu
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Guiwen Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Shuizhong Cen
- Department of Orthopedics, Zhujiang Hospital of Southern Medical Universuty, Guangzhou 510280, P.R. China
| | - Rujia Mi
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Xiaohua Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Wen Deng
- Center for Biotherapy, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Pei Feng
- Center for Biotherapy, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, P.R. China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, P.R. China
| |
Collapse
|
8
|
Kabatas S, Civelek E, Savrunlu EC, Kaplan N, Boyalı O, Diren F, Can H, Genç A, Akkoç T, Karaöz E. Feasibility of allogeneic mesenchymal stem cells in pediatric hypoxic-ischemic encephalopathy: Phase I study. World J Stem Cells 2021; 13:470-484. [PMID: 34136076 PMCID: PMC8176840 DOI: 10.4252/wjsc.v13.i5.470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/26/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is one of the leading causes of death and long-term neurological impairment in the pediatric population. Despite a limited number of treatments to cure HIE, stem cell therapies appear to be a potential treatment option for brain injury resulting from HIE.
AIM To investigate the efficacy and safety of stem cell-based therapies in pediatric patients with HIE.
METHODS The study inclusion criteria were determined as the presence of substantial deficit and disability caused by HIE. Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) were intrathecally (IT), intramuscularly (IM), and intravenously administered to participants at a dose of 1 × 106/kg for each administration route twice monthly for 2 mo. In different follow-up durations, the effect of WJ-MSCs administration on HIE, the quality of life, prognosis of patients, and side effects were investigated, and patients were evaluated for neurological, cognitive functions, and spasticity using the Wee Functional Independence Measure (Wee FIM) Scale and Modified Ashworth (MA) Scale.
RESULTS For all participants (n = 6), the mean duration of exposure to hypoxia was 39.17 + 18.82 min, the mean time interval after HIE was 21.83 ± 26.60 mo, the mean baseline Wee FIM scale score was 13.5 ± 0.55, and the mean baseline MA scale score was 35 ± 9.08. Three patients developed only early complications such as low-grade fever, mild headache associated with IT injection, and muscle pain associated with IM injection, all of which were transient and disappeared within 24 h. The treatment was evaluated to be safe and effective as demonstrated by magnetic resonance imaging examinations, electroencephalographies, laboratory tests, and neurological and functional scores of patients. Patients exhibited significant improvements in all neurological functions through a 12-mo follow-up. The mean Wee FIM scale score of participants increased from 13.5 ± 0.55 to 15.17 ± 1.6 points (mean ± SD) at 1 mo (z = - 1.826, P = 0.068) and to 23.5 ± 3.39 points at 12 mo (z = -2.207, P = 0.027) post-treatment. The percentage of patients who achieved an excellent functional improvement (Wee FIM scale total score = 126) increased from 10.71% (at baseline) to 12.03% at 1 mo and to 18.65% at 12 mo post-treatment.
CONCLUSION Both the triple-route and multiple WJ-MSC implantations were safe and effective in pediatric patients with HIE with significant neurological and functional improvements. The results of this study support conducting further randomized, placebo-controlled studies on this treatment in the pediatric population.
Collapse
Affiliation(s)
- Serdar Kabatas
- Department of Neurosurgery, University of Health Sciences, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Turkey
- Pediatric Allergy-Immunology, Marmara University, Institute of Health Sciences, Istanbul 34854, Turkey
- Center for Stem Cell & Gene Therapy Research and Practice, University of Health Sciences, Istanbul 34255, Turkey
| | - Erdinç Civelek
- Department of Neurosurgery, University of Health Sciences, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Turkey
- Pediatric Allergy-Immunology, Marmara University, Institute of Health Sciences, Istanbul 34854, Turkey
| | - Eyüp Can Savrunlu
- Department of Neurosurgery, University of Health Sciences, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Turkey
| | - Necati Kaplan
- Department of Neurosurgery, Istanbul Rumeli University, Çorlu Reyap Hospital, Tekirdağ 59860, Turkey
| | - Osman Boyalı
- Department of Neurosurgery, University of Health Sciences, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Turkey
| | - Furkan Diren
- Department of Neurosurgery, University of Health Sciences, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Turkey
| | - Halil Can
- Department of Neurosurgery, Istanbul Biruni University, Faculty of Medicine, Istanbul 34010, Turkey
- Department of Neurosurgery, Istanbul Medicine Hospital, Istanbul 34203, Turkey
| | - Ali Genç
- Department of Neurosurgery, Istanbul Asya Hospital, Istanbul 34250, Turkey
| | - Tunç Akkoç
- Pediatric Allergy-Immunology, Marmara University, Istanbul 34899, Turkey
| | - Erdal Karaöz
- Center for Regenerative Medicine and Stem Cell Research & Manufacturing (LivMedCell), Liv Hospital, Istanbul 34340, Turkey
- Department of Histology and Embryology, Istinye University, Faculty of Medicine, Istanbul 34010, Turkey
- Center for Stem Cell and Tissue Engineering Research and Practice, Istinye University, Istanbul 34340, Turkey
| |
Collapse
|
9
|
Primary Cilia as a Biomarker in Mesenchymal Stem Cells Senescence: Influencing Osteoblastic Differentiation Potency Associated with Hedgehog Signaling Regulation. Stem Cells Int 2021; 2021:8850114. [PMID: 33574852 PMCID: PMC7857927 DOI: 10.1155/2021/8850114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
Bone tissue engineering-based therapy for bone lesions requires the expansion of seeding cells, such as autologous mesenchymal stem cells (MSCs). A major obstacle to this process is the loss of the phenotype and differentiation capacity of MSCs subjected to passage. Recent studies have suggested that primary cilia, primordial organelles that transduce multiple signals, particularly hedgehog signals, play a role in senescence. Therefore, we explored the relationships among senescence, primary cilia, and hedgehog signaling in MSCs. Ageing of MSCs by expansion in vitro was accompanied by increased cell doubling time. The osteogenic capacity of aged MSCs at passage 4 was compromised compared to that of primary cells. P4 MSCs exhibited reductions in the frequency and length of primary cilia associated with decreased intensity of Arl13b staining on cilia. Senescence also resulted in downregulation of the expression of hedgehog components and CDKN2A. Suppression of ciliogenesis reduced the gene expression of both Gli1, a key molecule in the hedgehog signaling pathway and ALP, a marker of osteoblastic differentiation. This study demonstrated that the senescence of MSCs induced the loss of osteoblastic differentiation potency and inactivated hedgehog signaling associated with attenuated ciliogenesis, indicating that primary cilia play a mediating role in and are biomarkers of MSC senescence; thus, future antisenescence strategies involving manipulation of primary cilia could be developed.
Collapse
|
10
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Cochrane Database Syst Rev 2020; 8:CD013202. [PMID: 32813884 PMCID: PMC7438027 DOI: 10.1002/14651858.cd013202.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy (HIE) is a leading cause of mortality and long-term neurological sequelae, affecting thousands of children worldwide. Current therapies to treat HIE are limited to cooling. Stem cell-based therapies offer a potential therapeutic approach to repair or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal trials. OBJECTIVES To determine the efficacy and safety of stem cell-based interventions for the treatment of hypoxic-ischaemic encephalopathy (HIE) in newborn infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 5), MEDLINE via PubMed (1966 to 8 June 2020), Embase (1980 to 8 June 2020), and CINAHL (1982 to 8 June 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Randomised controlled trials, quasi-randomised controlled trials and cluster trials comparing 1) stem cell-based interventions (any type) compared to control (placebo or no treatment); 2) use of mesenchymal stem/stromal cells (MSCs) of type (e.g. number of doses or passages) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus MSCs of other type or source; 3) use of stem cell-based interventions other than MSCs of type (e.g. mononuclear cells, oligodendrocyte progenitor cells, neural stem cells, hematopoietic stem cells, and inducible pluripotent stem cells) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus stem cell-based interventions other than MSCs of other type or source; or 4) MSCs versus stem cell-based interventions other than MSCs. DATA COLLECTION AND ANALYSIS For each of the included trials, two authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs or other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). The primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, death or major neurodevelopmental disability assessed at 18 to 24 months of age. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 616 references. Two review authors independently assessed all references for inclusion. We did not find any completed studies for inclusion. Fifteen RCTs are currently registered and ongoing. We describe the three studies we excluded. AUTHORS' CONCLUSIONS There is currently no evidence from randomised trials that assesses the benefit or harms of stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
11
|
Romantsik O, Bruschettini M, Moreira A, Thébaud B, Ley D. Stem cell-based interventions for the prevention and treatment of germinal matrix-intraventricular haemorrhage in preterm infants. Cochrane Database Syst Rev 2019; 9:CD013201. [PMID: 31549743 PMCID: PMC6757514 DOI: 10.1002/14651858.cd013201.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Germinal matrix-intraventricular haemorrhage (GMH-IVH) remains a substantial issue in neonatal intensive care units worldwide. Current therapies to prevent or treat GMH-IVH are limited. Stem cell-based therapies offer a potential therapeutic approach to repair, restore, and/or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal studies. OBJECTIVES To determine the benefits and harms of stem cell-based interventions for prevention or treatment of germinal matrix-intraventricular haemorrhage (GM-IVH) in preterm infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2019, Issue 1), in the Cochrane Library; MEDLINE via PubMed (1966 to 7 January 2019); Embase (1980 to 7 January 2019); and the Cumulative Index to Nursing and Allied Health Literature (CINAHL) (1982 to 7 January 2019). We also searched clinical trials databases, conference proceedings, and reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA We attempted to identify randomised controlled trials, quasi-randomised controlled trials, and cluster trials comparing (1) stem cell-based interventions versus control; (2) mesenchymal stromal cells (MSCs) of type or source versus MSCs of other type or source; (3) stem cell-based interventions other than MSCs of type or source versus stem cell-based interventions other than MSCs of other type or source; or (4) MSCs versus stem cell-based interventions other than MSCs. For prevention studies, we included extremely preterm infants (less than 28 weeks' gestation), 24 hours of age or less, without ultrasound diagnosis of GM-IVH; for treatment studies, we included preterm infants (less than 37 weeks' gestation), of any postnatal age, with ultrasound diagnosis of GM-IVH. DATA COLLECTION AND ANALYSIS For each of the included trials, two review authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs, other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). Primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, GM-IVH, and extension of pre-existing non-severe GM-IVH. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 769 references. We did not find any completed studies for inclusion. One randomised controlled trial is currently registered and ongoing. Five phase 1 trials are described in the excluded studies. AUTHORS' CONCLUSIONS Currently no evidence is available to show the benefits or harms of stem cell-based interventions for treatment or prevention of GM-IVH in preterm infants.
Collapse
Affiliation(s)
- Olga Romantsik
- Lund University, Skåne University HospitalDepartment of Clinical Sciences Lund, PaediatricsLundSweden
| | - Matteo Bruschettini
- Lund University, Skåne University HospitalDepartment of Clinical Sciences Lund, PaediatricsLundSweden
- Skåne University HospitalCochrane SwedenWigerthuset, Remissgatan 4, first floorroom 11‐221LundSweden22185
| | - Alvaro Moreira
- University of Texas Health Science Center at San AntonioPediatrics, Division of NeonatologySan AntonioTexasUSA
| | - Bernard Thébaud
- Children's Hospital of Eastern OntarioDepartment of PediatricsOttawaONCanada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell ResearchOttawaCanada
- University of OttawaDepartment of Cellular and Molecular MedicineOttawaCanada
| | - David Ley
- Lund University, Skane University HospitalDepartment of Clinical Sciences Lund, PaediatricsLundSweden
| | | |
Collapse
|
12
|
Zhai W, Yong D, El-Jawhari JJ, Cuthbert R, McGonagle D, Win Naing M, Jones E. Identification of senescent cells in multipotent mesenchymal stromal cell cultures: Current methods and future directions. Cytotherapy 2019; 21:803-819. [PMID: 31138507 DOI: 10.1016/j.jcyt.2019.05.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/30/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
Regardless of their tissue of origin, multipotent mesenchymal stromal cells (MSCs) are commonly expanded in vitro for several population doublings to achieve a sufficient number of cells for therapy. Prolonged MSC expansion has been shown to result in phenotypical, morphological and gene expression changes in MSCs, which ultimately lead to the state of senescence. The presence of senescent cells in therapeutic MSC batches is undesirable because it reduces their viability, differentiation potential and trophic capabilities. Additionally, senescent cells acquire senescence-activated secretory phenotype, which may not only induce apoptosis in the neighboring host cells following MSC transplantation, but also trigger local inflammatory reactions. This review outlines the current and promising new methodologies for the identification of senescent cells in MSC cultures, with a particular emphasis on non-destructive and label-free methodologies. Technologies allowing identification of individual senescent cells, based on new surface markers, offer potential advantage for targeted senescent cell removal using new-generation senolytic agents, and subsequent production of therapeutic MSC batches fully devoid of senescent cells. Methods or a combination of methods that are non-destructive and label-free, for example, involving cell size and spectroscopic measurements, could be the best way forward because they do not modify the cells of interest, thus maximizing the final output of therapeutic-grade MSC cultures. The further incorporation of machine learning methods has also recently shown promise in facilitating, automating and enhancing the analysis of these measured data.
Collapse
Affiliation(s)
- Weichao Zhai
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK; Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Derrick Yong
- Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Jehan Jomaa El-Jawhari
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK; Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Richard Cuthbert
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK
| | - May Win Naing
- Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Elena Jones
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK.
| |
Collapse
|
13
|
Wiese DM, Ruttan CC, Wood CA, Ford BN, Braid LR. Accumulating Transcriptome Drift Precedes Cell Aging in Human Umbilical Cord-Derived Mesenchymal Stromal Cells Serially Cultured to Replicative Senescence. Stem Cells Transl Med 2019; 8:945-958. [PMID: 30924318 DOI: 10.1002/sctm.18-0246] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
In preclinical studies, mesenchymal stromal cells (MSCs) exhibit robust potential for numerous applications. To capitalize on these benefits, cell manufacturing and delivery protocols have been scaled up to facilitate clinical trials without adequately addressing the impact of these processes on cell utility nor inevitable regulatory requirements for consistency. Growing evidence indicates that culture-aged MSCs, expanded to the limits of replicative exhaustion to generate human doses, are not equivalent to early passage cells, and their use may underpin reportedly underwhelming or inconsistent clinical outcomes. Here, we sought to define the maximum expansion boundaries for human umbilical cord-derived MSCs, cultured in chemically defined xeno- and serum-free media, that yield consistent cell batches comparable to early passage cells. Two male and two female donor populations, recovered from cryostorage at mean population doubling level (mPDL) 10, were serially cultivated until replicative exhaustion (senescence). At each passage, growth kinetics, cell morphology, and transcriptome profiles were analyzed. All MSC populations displayed comparable growth trajectories through passage 9 (P9; mPDL 45) and variably approached senescence after P10 (mPDL 49). Transcription profiles of 14,500 human genes, generated by microarray, revealed a nonlinear evolution of culture-adapted MSCs. Significant expression changes occurred only after P5 (mPDL 27) and accumulated rapidly after P9 (mPDL 45), preceding other cell aging metrics. We report that cryobanked umbilical cord-derived MSCs can be reliably expanded to clinical human doses by P4 (mPDL 23), before significant transcriptome drift, and thus represent a mesenchymal cell source suited for clinical translation of cellular therapies. Stem Cells Translational Medicine 2019;8:945&958.
Collapse
Affiliation(s)
| | | | | | - Barry N Ford
- Casualty Management Section, DRDC Suffield Research Centre, Medicine Hat, Alberta, Canada
| | | |
Collapse
|
14
|
Marklein RA, Klinker MW, Drake KA, Polikowsky HG, Lessey-Morillon EC, Bauer SR. Morphological profiling using machine learning reveals emergent subpopulations of interferon-γ-stimulated mesenchymal stromal cells that predict immunosuppression. Cytotherapy 2018; 21:17-31. [PMID: 30503100 DOI: 10.1016/j.jcyt.2018.10.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/27/2018] [Accepted: 10/19/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although a preponderance of pre-clinical data demonstrates the immunosuppressive potential of mesenchymal stromal cells (MSCs), significant heterogeneity and lack of critical quality attributes (CQAs) based on immunosuppressive capacity likely have contributed to inconsistent clinical outcomes. This heterogeneity exists not only between MSC lots derived from different donors, tissues and manufacturing conditions, but also within a given MSC lot in the form of functional subpopulations. We therefore explored the potential of functionally relevant morphological profiling (FRMP) to identify morphological subpopulations predictive of the immunosuppressive capacity of MSCs derived from multiple donors, manufacturers and passages. METHODS We profiled the single-cell morphological response of MSCs from different donors and passages to the functionally relevant inflammatory cytokine interferon (IFN)-γ. We used the machine learning approach visual stochastic neighbor embedding (viSNE) to identify distinct morphological subpopulations that could predict suppression of activated CD4+ and CD8+ T cells in a multiplexed quantitative assay. RESULTS Multiple IFN-γ-stimulated subpopulations significantly correlated with the ability of MSCs to inhibit CD4+ and CD8+ T-cell activation and served as effective CQAs to predict the immunosuppressive capacity of additional manufactured MSC lots. We further characterized the emergence of morphological heterogeneity following IFN-γ stimulation, which provides a strategy for identifying functional subpopulations for future single-cell characterization and enrichment techniques. DISCUSSION This work provides a generalizable analytical platform for assessing functional heterogeneity based on single-cell morphological responses that could be used to identify novel CQAs and inform cell manufacturing decisions.
Collapse
Affiliation(s)
- Ross A Marklein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA; School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA.
| | - Matthew W Klinker
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Elizabeth C Lessey-Morillon
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
15
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| |
Collapse
|
16
|
Romantsik O, Bruschettini M, Moreira A, Thébaud B, Ley D. Stem cell-based interventions for the prevention and treatment of germinal matrix-intraventricular haemorrhage in preterm infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| |
Collapse
|
17
|
Shen C, Jiang T, Zhu B, Le Y, Liu J, Qin Z, Chen H, Zhong G, Zheng L, Zhao J, Zhang X. In vitro culture expansion impairs chondrogenic differentiation and the therapeutic effect of mesenchymal stem cells by regulating the unfolded protein response. J Biol Eng 2018; 12:26. [PMID: 30479659 PMCID: PMC6245887 DOI: 10.1186/s13036-018-0119-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023] Open
Abstract
In vitro expansion of mesenchymal stem cells (MSCs) has been implicated in loss of multipotency, leading to impaired chondrogenic potential and an eventual therapeutic effect, as reported in our previous study. However, the precise regulatory mechanism is still unclear. Here, we demonstrate that endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) were involved in transformation of MSCs induced by in vitro culture based on the comparative profiling of in vitro cultured bone marrow MSCs at passage 3 (P3 BMSCs) vs. fresh P0 BMSCs by microarray analysis. Indeed, RT-PCR and Western blot analysis showed significantly lower expression levels of three key UPR-related molecules, ATF4, ATF6 and XBP1, in P3 BMSCs than P0 BMSCs. Further, we found that UPR suppression by 4-phenylbutyrate (4-PBA) reduced the chondrogenic potential of P0 BMSCs and further cartilage regeneration. Conversely, UPR induction by tunicamycin (TM) enhanced the chondrogenic differentiation of P3 BMSCs and the therapeutic effect on cartilage repair. Thus, the decline in the chondrogenic potential of stem cells after in vitro culture and expansion may be due to changes in ER stress and the UPR pathway.
Collapse
Affiliation(s)
- Chong Shen
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China.,2Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tongmeng Jiang
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China.,2Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bo Zhu
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China
| | - Yiguan Le
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China.,2Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianwei Liu
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China
| | - Zainen Qin
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China
| | - Haimin Chen
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China
| | - Gang Zhong
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China.,2Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Zheng
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China
| | - Jinmin Zhao
- 1Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021 China.,2Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,3Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Xingdong Zhang
- 4National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064 China
| |
Collapse
|
18
|
In Vitro Expansion and Characterization of Mesenchymal Stromal Cells from Peritoneal Dialysis Effluent in a Human Protein Medium. Stem Cells Int 2018; 2018:5868745. [PMID: 30402111 PMCID: PMC6192083 DOI: 10.1155/2018/5868745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/23/2018] [Accepted: 09/03/2018] [Indexed: 12/19/2022] Open
Abstract
The therapeutic potential of mesenchymal stromal cells (MSCs) from various tissue origins have extensively been explored in both experimental and clinical studies, and peritoneal dialysis effluent-derived MSC (pMSC) may be an easily obtainable MSC source for clinical applications. In this study, we expanded and characterized the pMSCs after expansion in a human protein culture medium. The pMSCs were expanded in plastic dishes with the human protein medium. MSC marker expression was examined by flow cytometry. Spherical formation was tested by hanging drop method, and osteogenic, adipogenic, and chondrogenic differentiation capacities were confirmed by positive staining with Alizarin red, Oil red O, and Alcian blue, respectively. Here, we showed that after four passages of culturing in plastic dishes, pMSCs in the human protein medium displayed a homogeneous pattern of classical MSC markers (positive: CD29, CD44, CD73, CD90, and CD166; negative: CD14, CD34, CD45, CD79a, CD105, CD146, CD271, HLA-DR, SSEA-4, and Stro-1), while in the standard medium, pMSCs from some donors were CD45 or HLA-DR positive. For nonclassical MSC markers, pMSCs were CD200 positive from all the donors, negative for CD163, CD271, CD36, and CD248, and either positive or negative for CD274 and CD140b. Further, pMSCs from the human protein medium had the spherical formation capacity and multipotent differentiation capacity in vitro. In conclusion, upon expansion in a human protein medium, pMSCs showed a differential MSC marker expression profile from those of bone marrow or adipose tissue-derived MSCs and could maintain the multipotency. The therapeutic potential of the pMSCs requires further investigation.
Collapse
|
19
|
Lam J, Bellayr IH, Marklein RA, Bauer SR, Puri RK, Sung KE. Functional Profiling of Chondrogenically Induced Multipotent Stromal Cell Aggregates Reveals Transcriptomic and Emergent Morphological Phenotypes Predictive of Differentiation Capacity. Stem Cells Transl Med 2018; 7:664-675. [PMID: 30084545 PMCID: PMC6127231 DOI: 10.1002/sctm.18-0065] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/07/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Multipotent stromal cells (MSCs) are an attractive cell source for bone and cartilage tissue repair strategies. However, the functional heterogeneity of MSCs derived from different donors and manufacturing conditions has limited clinical translation, emphasizing the need for improved methods to assess MSC chondrogenic capacity. We used functionally relevant morphological profiling to dynamically monitor emergent morphological phenotypes of chondrogenically induced MSC aggregates to identify morphological features indicative of MSC chondrogenesis. Toward this goal, we characterized the morphology of chondrogenically stimulated MSC aggregates from eight different human cell-lines at multiple passages and demonstrated that MSC aggregates exhibited unique morphological dynamics that were both cell line- and passage-dependent. This variation in 3D morphology was shown to be informative of long-term MSC chondrogenesis based on multiple quantitative functional assays. We found that the specific morphological features of spheroid area, radius, minimum feret diameter, and minor axis length to be strongly correlated with MSC chondrogenic synthetic activity but not gene expression as early as day 4 in 3D culture. Our high-throughput, nondestructive approach could potentially serve as a tool to identify MSC lines with desired chondrogenic capacity toward improving manufacturing strategies for MSC-based cellular products for cartilage tissue repair. Stem Cells Translational Medicine 2018;1-12.
Collapse
Affiliation(s)
- Johnny Lam
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ian H Bellayr
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ross A Marklein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Raj K Puri
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kyung E Sung
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
20
|
Arcidiacono JA, Bauer SR, Kaplan DS, Allocca CM, Sarkar S, Lin-Gibson S. FDA and NIST collaboration on standards development activities supporting innovation and translation of regenerative medicine products. Cytotherapy 2018; 20:779-784. [PMID: 29784433 DOI: 10.1016/j.jcyt.2018.03.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/30/2018] [Indexed: 02/04/2023]
Abstract
The development of standards for the field of regenerative medicine has been noted as a high priority by several road-mapping activities. Additionally, the U.S. Congress recognizes the importance of standards in the 21st Century Cure Act. Standards will help to accelerate and streamline cell and gene therapy product development, ensure the quality and consistency of processes and products, and facilitate their regulatory approval. Although there is general agreement for the need of additional standards for regenerative medicine products, a shared understanding of standards is required for real progress toward the development of standards to advance regenerative medicine. Here, we describe the roles of standards in regenerative medicine as well as the process for standards development and the interactions of different entities in the standards development process. Highlighted are recent coordinated efforts between the U.S. Food and Drug Administration and the National Institute of Standards and Technology to facilitate standards development and foster science that underpins standards development.
Collapse
Affiliation(s)
- Judith A Arcidiacono
- Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Steven R Bauer
- Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - David S Kaplan
- Office of Science and Engineering Laboratory, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD USA
| | - Clare M Allocca
- Standards Coordination Office, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Sumona Sarkar
- Biosystems and Biomaterials Division, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Sheng Lin-Gibson
- Biosystems and Biomaterials Division, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, USA
| |
Collapse
|
21
|
Kundrotas G, Gasperskaja E, Slapsyte G, Gudleviciene Z, Krasko J, Stumbryte A, Liudkeviciene R. Identity, proliferation capacity, genomic stability and novel senescence markers of mesenchymal stem cells isolated from low volume of human bone marrow. Oncotarget 2017; 7:10788-802. [PMID: 26910916 PMCID: PMC4905439 DOI: 10.18632/oncotarget.7456] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/05/2016] [Indexed: 12/16/2022] Open
Abstract
Human bone marrow mesenchymal stem cells (hBM-MSCs) hold promise for treating incurable diseases and repairing of damaged tissues. However, hBM-MSCs face the disadvantages of painful invasive isolation and limited cell numbers. In this study we assessed characteristics of MSCs isolated from residual human bone marrow transplantation material and expanded to clinically relevant numbers at passages 3-4 and 6-7. Results indicated that early passage hBM-MSCs are genomically stable and retain identity and high proliferation capacity. Despite the chromosomal stability, the cells became senescent at late passages, paralleling the slower proliferation, altered morphology and immunophenotype. By qRT-PCR array profiling, we revealed 13 genes and 33 miRNAs significantly differentially expressed in late passage cells, among which 8 genes and 30 miRNAs emerged as potential novel biomarkers of hBM-MSC aging. Functional analysis of genes with altered expression showed strong association with biological processes causing cellular senescence. Altogether, this study revives hBM as convenient source for cellular therapy. Potential novel markers provide new details for better understanding the hBM-MSC senescence mechanisms, contributing to basic science, facilitating the development of cellular therapy quality control, and providing new clues for human disease processes since senescence phenotype of the hematological patient hBM-MSCs only very recently has been revealed.
Collapse
Affiliation(s)
- Gabrielis Kundrotas
- Department of Botany and Genetics, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania.,Biobank, National Cancer Institute, Vilnius, Lithuania
| | - Evelina Gasperskaja
- Department of Botany and Genetics, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania
| | - Grazina Slapsyte
- Department of Botany and Genetics, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania
| | | | - Jan Krasko
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania
| | | | | |
Collapse
|
22
|
Pierro M, Thébaud B, Soll R. Mesenchymal stem cells for the prevention and treatment of bronchopulmonary dysplasia in preterm infants. Cochrane Database Syst Rev 2017; 11:CD011932. [PMID: 29125893 PMCID: PMC6485972 DOI: 10.1002/14651858.cd011932.pub2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) remains a major complication of prematurity and currently lacks efficient treatments. Mesenchymal stem/stromal cells (MSCs) have been extensively explored as a potential therapy in several preclinical and clinical settings. Human and animal MSCs have been shown to prevent and treat lung injury in various preclinical models of lung diseases, including experimental BPD. OBJECTIVES To determine if MSCs, administered intravenously or endotracheally, are safe and effective in preventing or treating BPD, or both, in preterm infants. SEARCH METHODS We used the standard search strategy of the Cochrane Neonatal Review Group to search the Cochrane Central Register of Controlled Trials (CENTRAL 2016, Issue 10), MEDLINE via PubMed (1966 to 6 November 2016), Embase (1980 to 6 November 2016), and CINAHL (1982 to 6 November 2016). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials (RCTs) and quasi-RCTs. SELECTION CRITERIA We considered RCTs and quasi-RCTs investigating prevention or treatment of BPD, or both, in preterm infants. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trial quality according to prespecified criteria. MAIN RESULTS We found no RCTs or quasi-RCTs addressing the use of MSCs for prevention or treatment of BPD in premature infants. Two RCTs are currently registered and ongoing. AUTHORS' CONCLUSIONS There is insufficient evidence to determine the safety and efficacy of MSCs in the treatment or prevention of BPD in premature infants. The results of the ongoing trials addressing this issue are expected in the near future.
Collapse
Affiliation(s)
- Maria Pierro
- University of Milan, Fondazione IRCCS Cà Granda Ospedale Maggiore PoliclinicoDepartment of Clinical Sciences and Community HealthMilanItaly
- Alessandro Manzoni HospitalNeonatal Intensive Care UnitLeccoItaly
| | - Bernard Thébaud
- Children’s Hospital of Eastern OntarioDepartment of PediatricsOttawaONCanada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell ResearchOttawaCanada
- University of OttawaDepartment of Cellular and Molecular MedicineOttawaCanada
| | - Roger Soll
- University of Vermont Medical CenterDivision of Neonatal‐Perinatal Medicine111 Colchester AvenueBurlingtonVermontUSA05401
| | | |
Collapse
|
23
|
Lam J, Marklein RA, Jimenez-Torres JA, Beebe DJ, Bauer SR, Sung KE. Adaptation of a Simple Microfluidic Platform for High-Dimensional Quantitative Morphological Analysis of Human Mesenchymal Stromal Cells on Polystyrene-Based Substrates. SLAS Technol 2017; 22:646-661. [PMID: 28825968 DOI: 10.1177/2472630317726050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multipotent stromal cells (MSCs, often called mesenchymal stem cells) have garnered significant attention within the field of regenerative medicine because of their purported ability to differentiate down musculoskeletal lineages. Given the inherent heterogeneity of MSC populations, recent studies have suggested that cell morphology may be indicative of MSC differentiation potential. Toward improving current methods and developing simple yet effective approaches for the morphological evaluation of MSCs, we combined passive pumping microfluidic technology with high-dimensional morphological characterization to produce robust tools for standardized high-throughput analysis. Using ultraviolet (UV) light as a modality for reproducible polystyrene substrate modification, we show that MSCs seeded on microfluidic straight channel devices incorporating UV-exposed substrates exhibited morphological changes that responded accordingly to the degree of substrate modification. Substrate modification also effected greater morphological changes in MSCs seeded at a lower rather than higher density within microfluidic channels. Despite largely comparable trends in morphology, MSCs seeded in microscale as opposed to traditional macroscale platforms displayed much higher sensitivity to changes in substrate properties. In summary, we adapted and qualified microfluidic cell culture platforms comprising simple straight channel arrays as a viable and robust tool for high-throughput quantitative morphological analysis to study cell-material interactions.
Collapse
Affiliation(s)
- Johnny Lam
- 1 Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Ross A Marklein
- 1 Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Jose A Jimenez-Torres
- 2 Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- 2 Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven R Bauer
- 1 Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Kyung E Sung
- 1 Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
24
|
Bellayr IH, Kumar A, Puri RK. MicroRNA expression in bone marrow-derived human multipotent Stromal cells. BMC Genomics 2017; 18:605. [PMID: 28800721 PMCID: PMC5553681 DOI: 10.1186/s12864-017-3997-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022] Open
Abstract
Background Multipotent stromal cells (MSCs) are being studied in the field of regenerative medicine for their multi-lineage differentiation and immunoregulatory capacity. MicroRNAs (miRNAs) are short non-coding RNAs that are responsible for regulating gene expression by targeting transcripts, which can impact MSC functions such as cellular proliferation, differentiation, migration and cell death. miRNAs are expressed in MSCs; however, the impact of miRNAs on cellular functions and donor variability is not well understood. Eight MSC lines were expanded to passages 3, 5 and 7, and their miRNA expression was evaluated using microarray technology. Results Statistical analyses of our data revealed that 71 miRNAs out of 939 examined were expressed by this set of MSC lines at all passages and the expression of 11 miRNAs were significantly different between passages 3 and 7, while the expression of 7 miRNAs was significantly different between passages 3 and 5. The expression of these identified miRNAs was evaluated using RT-qPCR for both the first set of MSC lines (n = 6) and a second set of MSC lines (n = 7) expanded from passages 4 to 8. By RT-qPCR only 2 miRNAs, miR-638 and miR-572 were upregulated at passage 7 compared to passage 3 in the first set of MSC lines by 1.71 and 1.54 fold, respectively; and upregulated at passage 8 compared to passage 4 in the second set of MSC lines, 1.35 and 1.59 fold, respectively. Conclusions The expression of miR-638 and miR-572 can distinguish MSCs from two different passages of cell culture. These results may be useful in establishing critical quality attributes of MSCs and determining whether changes in these two miRNAs impact cellular functions. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3997-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ian H Bellayr
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics and Evaluation Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Abhinav Kumar
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics and Evaluation Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Raj K Puri
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics and Evaluation Research, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
25
|
MicroRNA profiling analysis revealed different cellular senescence mechanisms in human mesenchymal stem cells derived from different origin. Genomics 2017; 109:147-157. [DOI: 10.1016/j.ygeno.2017.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 01/01/2023]
|
26
|
DiCarlo AL, Tamarat R, Rios CI, Benderitter M, Czarniecki CW, Allio TC, Macchiarini F, Maidment BW, Jourdain JR. Cellular Therapies for Treatment of Radiation Injury: Report from a NIH/NIAID and IRSN Workshop. Radiat Res 2017; 188:e54-e75. [PMID: 28605260 DOI: 10.1667/rr14810.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In recent years, there has been increasing concern over the possibility of a radiological or nuclear incident occurring somewhere in the world. Intelligence agencies frequently report that terrorist groups and rogue nations are seeking to obtain radiological or nuclear weapons of mass destruction. In addition, there exists the real possibility that safety of nuclear power reactors could be compromised by natural (such as the tsunami and subsequent Fukushima accident in Japan in March, 2011) or accidental (Three Mile Island, 1979 and Chernobyl, 1986) events. Although progress has been made by governments around the world to prepare for these events, including the stockpiling of radiation countermeasures, there are still challenges concerning care of patients injured during a radiation incident. Because the deleterious and pathological effects of radiation are so broad, it is desirable to identify medical countermeasures that can have a beneficial impact on several tissues and organ systems. Cellular therapies have the potential to impact recovery and tissue/organ regeneration for both early and late complications of radiation exposure. These therapies, which could include stem or blood progenitor cells, mesenchymal stromal cells (MSCs) or cells derived from other tissues (e.g., endothelium or placenta), have shown great promise in treating other nonradiation injuries to and diseases of the bone marrow, skin, gastrointestinal tract, brain, lung and heart. To explore the potential use of these therapies in the treatment of victims after acute radiation exposure, the National Institute of Allergy and Infectious Diseases co-sponsored an international workshop in July, 2015 in Paris, France with the Institut de Radioprotection et de Sûreté Nucléaire. The workshop included discussions of data available from testing in preclinical models of radiation injury to different organs, logistics associated with the practical use of cellular therapies for a mass casualty incident, as well as international regulatory requirements for authorizing such drug products to be legally and readily used in such incidents. This report reviews the data presented, as well as key discussion points from the meeting.
Collapse
Affiliation(s)
- Andrea L DiCarlo
- a Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Radia Tamarat
- b Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | - Carmen I Rios
- a Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Marc Benderitter
- b Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | | | | | - Francesca Macchiarini
- e Previously -RNCP, DAIT, NIAID, NIH; now National Institute on Aging (NIA), NIH, Bethesda, Maryland
| | | | - Jean-Rene Jourdain
- b Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| |
Collapse
|
27
|
Jiang T, Xu G, Wang Q, Yang L, Zheng L, Zhao J, Zhang X. In vitro expansion impaired the stemness of early passage mesenchymal stem cells for treatment of cartilage defects. Cell Death Dis 2017; 8:e2851. [PMID: 28569773 PMCID: PMC5520885 DOI: 10.1038/cddis.2017.215] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 02/08/2023]
Abstract
In vitro cultured autologous mesenchymal stem cells (MSCs) within passage 5 have been approved for clinical application in stem cell-based treatment of cartilage defects. However, their chondrogenic potential has not yet been questioned or verified. In this study, the chondrogenic potential of bone marrow MSCs at passage 3 (P3 BMSCs) was investigated both in cartilage repair and in vitro, with freshly isolated bone marrow mononuclear cells (BMMNCs) as controls. The results showed that P3 BMSCs were inferior to BMMNCs not only in their chondrogenic differentiation ability but also as candidates for long-term repair of cartilage defects. Compared with BMMNCs, P3 BMSCs presented a decay in telomerase activity and a change in chromosomal morphology with potential anomalous karyotypes, indicating senescence. In addition, interindividual variability in P3 BMSCs is much higher than in BMMNCs, demonstrating genomic instability. Interestingly, remarkable downregulation in cell cycle, DNA replication and mismatch repair (MMR) pathways as well as in multiple genes associated with telomerase activity and chromosomal stability were found in P3 BMSCs. This result indicates that telomerase and chromosome anomalies might originate from expansion, leading to impaired stemness and pluripotency of stem cells. In vitro culture and expansion are not recommended for cell-based therapy, and fresh BMMNCs are the first choice.
Collapse
Affiliation(s)
- Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Guojie Xu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, China
| | - Lihui Yang
- School of Nursing, Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Collaborative Innovation Center of Guangxi Biological Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Collaborative Innovation Center of Guangxi Biological Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.,Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
28
|
Wong TY, Chang CH, Yu CH, Huang LLH. Hyaluronan keeps mesenchymal stem cells quiescent and maintains the differentiation potential over time. Aging Cell 2017; 16:451-460. [PMID: 28474484 PMCID: PMC5418204 DOI: 10.1111/acel.12567] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hyaluronan (HA), an abundant polysaccharide found in human bodies, plays a role in the mesenchymal stem cells (MSCs) maintenance. We had previously found that HA prolonged the lifespan, and prevented the cellular aging of murine adipose-derived stromal cells. Recently, we had also summarized the potential pathways associated with HA regulation in human MSCs. In this study, we used the human placenta-derived MSCs (PDMSC) to investigate the effectiveness of HA in maintaining the PDMSC. We found that coating the culture surface coated with 30 μg cm-2 of HA (C) led to cluster growth of PDMSC, and maintained a higher number of PDMSC in quiescence compared to those grown on the normal tissue culture surface (T). PDMSC were treated for either 4 (short-term) or 19 (long-term) consecutive passages. PDMSC which were treated with HA for 19 consecutive passages had reduced cell enlargement, preserved MSCs biomarker expressions and osteogenic potential when compared to those grown only on T. The PDMSC transferred to T condition after long-term HA treatment showed preserved replicative capability compared to those on only T. The telomerase activity of the HA-treated PDMSC was also higher than that of untreated PDMSC. These data suggested a connection between HA and MSC maintenance. We suggest that HA might be regulating the distribution of cytoskeletal proteins on cell spreading in the event of quiescence to preserve MSC stemness. Maintenance of MSCs stemness delayed cellular aging, leading to the anti-aging phenotype of PDMSC.
Collapse
Affiliation(s)
- Tzyy Yue Wong
- Institute of Biotechnology; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
| | - Chiung-Hsin Chang
- Department of Obstetrics and Gynecology; National Cheng Kung University; Tainan Taiwan
| | - Chen-Hsiang Yu
- Department of Obstetrics and Gynecology; National Cheng Kung University; Tainan Taiwan
| | - Lynn L. H. Huang
- Institute of Biotechnology; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
- Department of Biotechnology and Bioindustry Sciences; College of Bioscience and Biotechnology; National Cheng Kung University; Tainan Taiwan
- Institute of Clinical Medicine; College of Medicine; National Cheng Kung University; Tainan Taiwan
- Research Center of Excellence in Regenerative Medicine; National Cheng Kung University; Tainan Taiwan
- Advanced Optoelectronic Technology Center; National Cheng Kung University; Tainan Taiwan
| |
Collapse
|
29
|
Klinker MW, Marklein RA, Lo Surdo JL, Wei CH, Bauer SR. Morphological features of IFN-γ-stimulated mesenchymal stromal cells predict overall immunosuppressive capacity. Proc Natl Acad Sci U S A 2017; 114:E2598-E2607. [PMID: 28283659 PMCID: PMC5380055 DOI: 10.1073/pnas.1617933114] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stromal cell (MSC) lines can vary significantly in their functional characteristics, and the effectiveness of MSC-based therapeutics may be realized by finding predictive features associated with MSC function. To identify features associated with immunosuppressive capacity in MSCs, we developed a robust in vitro assay that uses principal-component analysis to integrate multidimensional flow cytometry data into a single measurement of MSC-mediated inhibition of T-cell activation. We used this assay to correlate single-cell morphological data with overall immunosuppressive capacity in a cohort of MSC lines derived from different donors and manufacturing conditions. MSC morphology after IFN-γ stimulation significantly correlated with immunosuppressive capacity and accurately predicted the immunosuppressive capacity of MSC lines in a validation cohort. IFN-γ enhanced the immunosuppressive capacity of all MSC lines, and morphology predicted the magnitude of IFN-γ-enhanced immunosuppressive activity. Together, these data identify MSC morphology as a predictive feature of MSC immunosuppressive function.
Collapse
Affiliation(s)
- Matthew W Klinker
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Ross A Marklein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Jessica L Lo Surdo
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Cheng-Hong Wei
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993
| |
Collapse
|
30
|
Tian Y, Xu Y, Xue T, Chen L, Shi B, Shu B, Xie C, Max Morandi M, Jaeblon T, Marymont JV, Dong Y. Notch activation enhances mesenchymal stem cell sheet osteogenic potential by inhibition of cellular senescence. Cell Death Dis 2017; 8:e2595. [PMID: 28151468 PMCID: PMC5386477 DOI: 10.1038/cddis.2017.2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/21/2016] [Accepted: 12/28/2016] [Indexed: 01/21/2023]
Abstract
Our previous studies have confirmed the therapeutic effects of mesenchymal stem cell (MSC) monolayer sheet transplantation on allograft repair. A limiting factor in their application is the loss of MSC multi-potency as a result of high density sheet culture-induced senescence. In the study reported in this article, we tested whether Notch activation could be used to prevent or delay sheet culture-induced cell aging. Our results showed that, during in vitro long-term (5-day) cell sheet culture, MSCs progressively lose their progenitor characteristics. In contrast, Notch activation by Jagged1 in MSC sheet culture showed reduced cellular senescence and cell cycle arrest compared with control MSCs without Notch activation. Importantly, knockdown of Notch target gene Hes1 totally blocked the inhibition effect of Jagged1 on cellular senescence. Finally, the in vivo allograft transplantation data showed a significant enhanced callus formation and biomechanical properties in Notch activation cultured long-term sheet groups when compared with long-term cultured sheet without Notch activation. Our results suggest that Notch activation by Jagged1 could be used to overcome the stem cell aging caused by high density sheet culture, thereby increasing the therapeutic potential of MSC sheets for tissue regeneration.
Collapse
Affiliation(s)
- Ye Tian
- Department of Orthopedics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Taiyang Xue
- Department of Orthopedics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Longgang Chen
- Department of Orthopedics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Bin Shi
- Department of Orthopedics, Shengjing Hospital, China Medical University, Shenyang, China
| | - Bing Shu
- Department of Orthopedics, Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Xie
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Massimo Max Morandi
- Department of Orthopedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Todd Jaeblon
- Department of Orthopedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - John V Marymont
- Department of Orthopedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Yufeng Dong
- Department of Orthopedic Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
31
|
Canine Adipose Derived Mesenchymal Stem Cells Transcriptome Composition Alterations: A Step towards Standardizing Therapeutic. Stem Cells Int 2017; 2017:4176292. [PMID: 28246532 PMCID: PMC5299202 DOI: 10.1155/2017/4176292] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/19/2016] [Accepted: 01/04/2017] [Indexed: 01/09/2023] Open
Abstract
Although canine adipose derived stem cells (cASCs) morphology characteristics and differentiation ability are well documented, transcriptome alterations of undifferentiated cASCs during ex vivo cultivation remain unknown. Here we demonstrate, for the first time, the transcriptome composition of isolated cASCs in undifferentiated state originating from six donors. Transcriptome changes were monitored during ex vivo cultivation between passage 3 (P3) and P5, which are mostly used in therapy. Influence of donors' age in given passage number on transcriptome composition was also investigated. Cultivation from P3 to P5 resulted in 16 differentially expressed genes with cooverexpression of pluripotency and self-renewal transcription factors genes SOX2 and POU5F1 dominant in old donors' cells. Furthermore, cASCs demonstrated upregulation of IL-6 in young and old donors' cells. In addition, ex vivo cultivation of cASCs revealed well-known morphological alterations accompanied with decrease in expression of CD90 and CD44 markers in P4 and higher monitored by flow cytometry and successful osteo- and chondrodifferentiation but inefficient adipodifferentiation in P3. Our results revealed the impact of ex vivo cultivation on nature of cells. Correlation of transcriptome changes with secretome composition is needed and its further impact on therapeutic potential of cASCs remains to be evaluated in clinical trials.
Collapse
|
32
|
Altman RB, Khuri N, Salit M, Giacomini KM. Unmet needs: Research helps regulators do their jobs. Sci Transl Med 2016; 7:315ps22. [PMID: 26606966 DOI: 10.1126/scitranslmed.aac4369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A plethora of innovative new medical products along with the need to apply modern technologies to medical-product evaluation has spurred seminal opportunities in regulatory sciences. Here, we provide eight examples of regulatory science research for diverse products. Opportunities abound, particularly in data science and precision health.
Collapse
Affiliation(s)
- Russ B Altman
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Natalia Khuri
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, CA 94305, USA. Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94143-2911, USA
| | - Marc Salit
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, CA 94305, USA. Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA 94143-2911, USA. Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
33
|
Hofer HR, Tuan RS. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res Ther 2016; 7:131. [PMID: 27612948 PMCID: PMC5016979 DOI: 10.1186/s13287-016-0394-0] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adult mesenchymal stem cells (MSCs) represent a subject of intense experimental and biomedical interest. Recently, trophic activities of MSCs have become the topic of a number of revealing studies that span both basic and clinical fields. In this review, we focus on recent investigations that have elucidated trophic mechanisms and shed light on MSC clinical efficacy relevant to musculoskeletal applications. Innate differences due to MSC sourcing may play a role in the clinical utility of isolated MSCs. Pain management, osteochondral, nerve, or blood vessel support by MSCs derived from both autologous and allogeneic sources have been examined. Recent mechanistic insights into the trophic activities of these cells point to ultimate regulation by nitric oxide, nuclear factor-kB, and indoleamine, among other signaling pathways. Classic growth factors and cytokines-such as VEGF, CNTF, GDNF, TGF-β, interleukins (IL-1β, IL-6, and IL-8), and C-C ligands (CCL-2, CCL-5, and CCL-23)-serve as paracrine control molecules secreted or packaged into extracellular vesicles, or exosomes, by MSCs. Recent studies have also implicated signaling by microRNAs contained in MSC-derived exosomes. The response of target cells is further regulated by their microenvironment, involving the extracellular matrix, which may be modified by MSC-produced matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs. Trophic activities of MSCs, either resident or introduced exogenously, are thus intricately controlled, and may be further fine-tuned via implant material modifications. MSCs are actively being investigated for the repair and regeneration of both osteochondral and other musculoskeletal tissues, such as tendon/ligament and meniscus. Future rational and effective MSC-based musculoskeletal therapies will benefit from better mechanistic understanding of MSC trophic activities, for example using analytical "-omics" profiling approaches.
Collapse
Affiliation(s)
- Heidi R Hofer
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
34
|
Senescence in Human Mesenchymal Stem Cells: Functional Changes and Implications in Stem Cell-Based Therapy. Int J Mol Sci 2016; 17:ijms17071164. [PMID: 27447618 PMCID: PMC4964536 DOI: 10.3390/ijms17071164] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/04/2016] [Accepted: 07/14/2016] [Indexed: 12/19/2022] Open
Abstract
Regenerative medicine is extensively interested in developing cell therapies using mesenchymal stem cells (MSCs), with applications to several aging-associated diseases. For successful therapies, a substantial number of cells are needed, requiring extensive ex vivo cell expansion. However, MSC proliferation is limited and it is quite likely that long-term culture evokes continuous changes in MSCs. Therefore, a substantial proportion of cells may undergo senescence. In the present review, we will first present the phenotypic characterization of senescent human MSCs (hMSCs) and their possible consequent functional alterations. The accumulation of oxidative stress and dysregulation of key differentiation regulatory factors determine decreased differentiation potential of senescent hMSCs. Senescent hMSCs also show a marked impairment in their migratory and homing ability. Finally, many factors present in the secretome of senescent hMSCs are able to exacerbate the inflammatory response at a systemic level, decreasing the immune modulation activity of hMSCs and promoting either proliferation or migration of cancer cells. Considering the deleterious effects that these changes could evoke, it would appear of primary importance to monitor the occurrence of senescent phenotype in clinically expanded hMSCs and to evaluate possible ways to prevent in vitro MSC senescence. An updated critical presentation of the possible strategies for in vitro senescence monitoring and prevention constitutes the second part of this review. Understanding the mechanisms that drive toward hMSC growth arrest and evaluating how to counteract these for preserving a functional stem cell pool is of fundamental importance for the development of efficient cell-based therapeutic approaches.
Collapse
|
35
|
Rovira Gonzalez YI, Lynch PJ, Thompson EE, Stultz BG, Hursh DA. In vitro cytokine licensing induces persistent permissive chromatin at the Indoleamine 2,3-dioxygenase promoter. Cytotherapy 2016; 18:1114-28. [PMID: 27421739 DOI: 10.1016/j.jcyt.2016.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 05/09/2016] [Accepted: 05/24/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are being investigated as therapies for inflammatory diseases due to their immunosuppressive capacity. Interferon (IFN)-γ treatment primes MSC immunosuppression partially through induction of Indoleamine 2,3-dioxygenase (IDO1), which depletes tryptophan necessary to support proliferation of activated T cells. We investigated the role of histone modifications in the timing and maintenance of induced IDO1 expression in MSCs under clinical manufacturing conditions, such as cryopreservation. METHODS We used chromatin immunoprecipitation and quantitative polymerase chain reaction (PCR) to assay levels of transcriptionally permissive acetylated H3K9 and repressive trimethylated H3K9 histone modifications surrounding the transcriptional start site for IDO1, and reverse transcriptase PCR and immunoblotting to detect messenger RNA (mRNA) and protein. RESULTS MSCs derived from three donors approached maximum IDO1 mRNA levels following 24 hours of in vitro cytokine treatment. Induction of IDO1 expression correlated with increased acetylation of H3K9 concomitant with reduction of trimethylated H3K9 modifications at the promoter. Examination of two additional donors confirmed this result. While induced IDO1 levels decreased within 2 days after cytokine removal and freeze thawing, the activated chromatin state was maintained. Upon re-exposure to cytokines, previously primed MSCs accumulated near-maximum IDO1 mRNA levels within 4-8 h. DISCUSSION Our data indicate that in vitro priming of MSCs causes chromatin remodeling at the IDO1 promoter, that this alteration is maintained during processing commonly used to prepare MSCs for clinical use and that, once primed, MSCs are poised for IDO1 expression even in the absence of cytokines.
Collapse
Affiliation(s)
- Yazmin I Rovira Gonzalez
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Patrick J Lynch
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.
| | - Elaine E Thompson
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Brian G Stultz
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Deborah A Hursh
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
36
|
Manufacturing Cells for Clinical Use. Stem Cells Int 2016; 2016:1750697. [PMID: 27382370 PMCID: PMC4921138 DOI: 10.1155/2016/1750697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/03/2016] [Indexed: 02/07/2023] Open
|
37
|
Bellayr IH, Marklein RA, Lo Surdo JL, Bauer SR, Puri RK. Identification of Predictive Gene Markers for Multipotent Stromal Cell Proliferation. Stem Cells Dev 2016; 25:861-73. [PMID: 27036644 DOI: 10.1089/scd.2015.0374] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multipotent stromal cells (MSCs) are known for their distinctive ability to differentiate into different cell lineages, such as adipocytes, chondrocytes, and osteocytes. They can be isolated from numerous tissue sources, including bone marrow, adipose tissue, skeletal muscle, and others. Because of their differentiation potential and secretion of growth factors, MSCs are believed to have an inherent quality of regeneration and immune suppression. Cellular expansion is necessary to obtain sufficient numbers for use; however, MSCs exhibit a reduced capacity for proliferation and differentiation after several rounds of passaging. In this study, gene markers of MSC proliferation were identified and evaluated for their ability to predict proliferative quality. Microarray data of human bone marrow-derived MSCs were correlated with two proliferation assays. A collection of 24 genes were observed to significantly correlate with both proliferation assays (|r| >0.70) for eight MSC lines at multiple passages. These 24 identified genes were then confirmed using an additional set of MSCs from eight new donors using reverse transcription quantitative polymerase chain reaction (RT-qPCR). The proliferative potential of the second set of MSCs was measured for each donor/passage for confluency fraction, fraction of EdU+ cells, and population doubling time. The second set of MSCs exhibited a greater proliferative potential at passage 4 in comparison to passage 8, which was distinguishable by 15 genes; however, only seven of the genes (BIRC5, CCNA2, CDC20, CDK1, PBK, PLK1, and SPC25) demonstrated significant correlation with MSC proliferation regardless of passage. Our analyses revealed that correlation between gene expression and proliferation was consistently reduced with the inclusion of non-MSC cell lines; therefore, this set of seven genes may be more strongly associated with MSC proliferative quality. Our results pave the way to determine the quality of an MSC population for a particular cellular therapy in lieu of an extended in vitro or in vivo assay.
Collapse
Affiliation(s)
- Ian H Bellayr
- 1 Division of Cellular and Gene Therapies, Tumor Vaccines and Biotechnology Branch, Center for Biologics and Evaluation Research , US Food and Drug Administration, Silver Spring, Maryland
| | - Ross A Marklein
- 2 Division of Cellular and Gene Therapies, Cellular and Tissue Therapies Branch, Center for Biologics Evaluation and Research , US Food and Drug Administration, Silver Spring, Maryland
| | - Jessica L Lo Surdo
- 2 Division of Cellular and Gene Therapies, Cellular and Tissue Therapies Branch, Center for Biologics Evaluation and Research , US Food and Drug Administration, Silver Spring, Maryland
| | - Steven R Bauer
- 2 Division of Cellular and Gene Therapies, Cellular and Tissue Therapies Branch, Center for Biologics Evaluation and Research , US Food and Drug Administration, Silver Spring, Maryland
| | - Raj K Puri
- 1 Division of Cellular and Gene Therapies, Tumor Vaccines and Biotechnology Branch, Center for Biologics and Evaluation Research , US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
38
|
Marklein RA, Lo Surdo JL, Bellayr IH, Godil SA, Puri RK, Bauer SR. High Content Imaging of Early Morphological Signatures Predicts Long Term Mineralization Capacity of Human Mesenchymal Stem Cells upon Osteogenic Induction. Stem Cells 2016; 34:935-47. [PMID: 26865267 DOI: 10.1002/stem.2322] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/30/2015] [Indexed: 01/05/2023]
Abstract
Human bone marrow-derived multipotent mesenchymal stromal cells, often referred to as mesenchymal stem cells (MSCs), represent an attractive cell source for many regenerative medicine applications due to their potential for multi-lineage differentiation, immunomodulation, and paracrine factor secretion. A major complication for current MSC-based therapies is the lack of well-defined characterization methods that can robustly predict how they will perform in a particular in vitro or in vivo setting. Significant advances have been made with identifying molecular markers of MSC quality and potency using multivariate genomic and proteomic approaches, and more recently with advanced techniques incorporating high content imaging to assess high-dimensional single cell morphological data. We sought to expand upon current methods of high dimensional morphological analysis by investigating whether short term cell and nuclear morphological profiles of MSCs from multiple donors (at multiple passages) correlated with long term mineralization upon osteogenic induction. Using the combined power of automated high content imaging followed by automated image analysis, we demonstrated that MSC morphology after 3 days was highly correlated with 35 day mineralization and comparable to other methods of MSC osteogenesis assessment (such as alkaline phosphatase activity). We then expanded on this initial morphological characterization and identified morphological features that were highly predictive of mineralization capacities (>90% accuracy) of MSCs from additional donors and different manufacturing techniques using linear discriminant analysis. Together, this work thoroughly demonstrates the predictive power of MSC morphology for mineralization capacity and motivates further studies into MSC morphology as a predictive marker for additional in vitro and in vivo responses.
Collapse
Affiliation(s)
- Ross A Marklein
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jessica L Lo Surdo
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ian H Bellayr
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Saniya A Godil
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Raj K Puri
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
39
|
Stultz BG, McGinnis K, Thompson EE, Lo Surdo JL, Bauer SR, Hursh DA. Chromosomal stability of mesenchymal stromal cells during in vitro culture. Cytotherapy 2016; 18:336-43. [PMID: 26780865 DOI: 10.1016/j.jcyt.2015.11.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/27/2015] [Accepted: 11/18/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are being investigated for use in cell therapy. The extensive in vitro expansion necessary to obtain sufficient cells for clinical use increases the risk that genetically abnormal cells will arise and be propagated during cell culture. Genetic abnormalities may lead to transformation and poor performance in clinical use, and are a critical safety concern for cell therapies using MSCs. METHODS We used spectral karyotyping (SKY) to investigate the genetic stability of human MSCs from ten donors during passaging. RESULTS Our data indicate that chromosomal abnormalities exist in MSCs at early passages and can be clonally propagated. The karyotypic abnormalities observed during our study diminished during passage. CONCLUSIONS Karyotyping of MSCs reveals characteristics which may be valuable in deciding the suitability of cells for further use. Karyotypic analysis is useful for monitoring the genetic stability of MSCs during expansion.
Collapse
Affiliation(s)
- Brian G Stultz
- Division of Cell and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kathleen McGinnis
- Division of Cell and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Elaine E Thompson
- Division of Cell and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jessica L Lo Surdo
- Division of Cell and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Division of Cell and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Deborah A Hursh
- Division of Cell and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
40
|
Mesenchymal stem cells for the prevention and treatment of bronchopulmonary dysplasia in preterm infants. Hippokratia 2015. [DOI: 10.1002/14651858.cd011932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
41
|
Lynch PJ, Thompson EE, McGinnis K, Rovira Gonzalez YI, Lo Surdo J, Bauer SR, Hursh DA. Chromatin Changes at thePPAR-γ2Promoter During Bone Marrow-Derived Multipotent Stromal Cell Culture Correlate With Loss of Gene Activation Potential. Stem Cells 2015; 33:2169-81. [DOI: 10.1002/stem.1967] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 01/06/2014] [Indexed: 01/01/2023]
Affiliation(s)
- Patrick J. Lynch
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| | - Elaine E. Thompson
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| | - Kathleen McGinnis
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| | - Yazmin I. Rovira Gonzalez
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| | - Jessica Lo Surdo
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| | - Steven R. Bauer
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| | - Deborah A. Hursh
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration; Bethesda Maryland USA
| |
Collapse
|
42
|
Tratwal J, Mathiasen AB, Juhl M, Brorsen SK, Kastrup J, Ekblond A. Influence of vascular endothelial growth factor stimulation and serum deprivation on gene activation patterns of human adipose tissue-derived stromal cells. Stem Cell Res Ther 2015; 6:62. [PMID: 25889587 PMCID: PMC4431456 DOI: 10.1186/s13287-015-0062-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 11/05/2014] [Accepted: 03/24/2015] [Indexed: 02/07/2023] Open
Abstract
Introduction Stimulation of mesenchymal stromal cells and adipose tissue-derived stromal cells (ASCs) with vascular endothelial growth factor (VEGF) has been used in multiple animal studies and clinical trials for regenerative purposes. VEGF stimulation is believed to promote angiogenesis and VEGF stimulation is usually performed under serum deprivation. Potential regenerative molecular mechanisms are numerous and the role of contributing factors is uncertain. The aim of the current study was to investigate the effect of in vitro serum deprivation and VEGF stimulation on gene expression patterns of ASCs. Methods Gene expressions of ASCs cultured in complete medium, ASCs cultured in serum-deprived medium and ASCs stimulated with VEGF in serum-deprived medium were compared. ASC characteristics according to criteria set by the International Society of Cellular Therapy were confirmed by flow cytometry. Microarray gene expressions were obtained using the Affymetrix HT HG-U133+ GeneChip®. Gene set enrichment analysis was performed using the Kyoto Encyclopedia of Genes and Genomes and gene ontology terms. Transcription of selected genes of interest was confirmed by quantitative PCR. Results Compared to ASCs in complete medium, 190 and 108 genes were significantly altered by serum deprivation and serum deprivation combined with VEGF, respectively. No significant differences in gene expression patterns between serum-deprived ASCs and serum-deprived ASCs combined with VEGF stimulation were found. Genes most prominently and significantly upregulated by both conditions were growth factors (IGF1, BMP6, PDGFD, FGF9), adhesion molecule CLSTN2, extracellular matrix-related proteins such as matricellular proteins SMOC2, SPON1 and ADAMTS12, and inhibitors of proliferation (JAG1). The most significantly downregulated genes included matrix metalloproteinases (MMP3, MMP1), and proliferation markers (CDKN3) and GREM2 (a BMP6 antagonist). Conclusion The decisive factor for the observed change in ASC gene expression proves to be serum starvation rather than VEGF stimulation. Changes in expression of growth factors, matricellular proteins and matrix metalloproteinases in concert, diverge from direct pro-angiogenic paracrine mechanisms as a primary consequence of the used protocol. In vitro serum starvation (with or without VEGF present) appears to favour cardioprotection, extracellular matrix remodelling and blood vessel maturation relevant for the late maturation phase in infarct healing.
Collapse
Affiliation(s)
- Josefine Tratwal
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Juliane Maries Vej 20, dept. 9302, Copenhagen, 2100, Denmark.
| | - Anders Bruun Mathiasen
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Juliane Maries Vej 20, dept. 9302, Copenhagen, 2100, Denmark.
| | - Morten Juhl
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Juliane Maries Vej 20, dept. 9302, Copenhagen, 2100, Denmark.
| | - Sonja Kim Brorsen
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Juliane Maries Vej 20, dept. 9302, Copenhagen, 2100, Denmark.
| | - Jens Kastrup
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Juliane Maries Vej 20, dept. 9302, Copenhagen, 2100, Denmark.
| | - Annette Ekblond
- Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Juliane Maries Vej 20, dept. 9302, Copenhagen, 2100, Denmark.
| |
Collapse
|
43
|
Impact of umbilical cord blood-derived mesenchymal stem cells on cardiovascular research. BIOMED RESEARCH INTERNATIONAL 2015; 2015:975302. [PMID: 25861654 PMCID: PMC4377460 DOI: 10.1155/2015/975302] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/11/2015] [Indexed: 01/06/2023]
Abstract
Over the years, cell therapy has become an exciting opportunity to treat human diseases. Early enthusiasm using adult stem cell sources has been tempered in light of preliminary benefits in patients. Considerable efforts have been dedicated, therefore, to explore alternative cells such as those extracted from umbilical cord blood (UCB). In line, UCB banking has become a popular possibility to preserve potentially life-saving cells that are usually discarded after birth, and the number of UCB banks has grown worldwide. Thus, a brief overview on the categories of UCB banks as well as the properties, challenges, and impact of UCB-derived mesenchymal stem cells (MSCs) on the area of cardiovascular research is presented. Taken together, the experience recounted here shows that UCBMSCs are envisioned as attractive therapeutic candidates against human disorders arising and/or progressing with vascular deficit.
Collapse
|
44
|
Baker N, Boyette LB, Tuan RS. Characterization of bone marrow-derived mesenchymal stem cells in aging. Bone 2015; 70:37-47. [PMID: 25445445 DOI: 10.1016/j.bone.2014.10.014] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 12/17/2022]
Abstract
Adult mesenchymal stem cells are a resource for autologous and allogeneic cell therapies for immune-modulation and regenerative medicine. However, patients most in need of such therapies are often of advanced age. Therefore, the effects of the aged milieu on these cells and their intrinsic aging in vivo are important considerations. Furthermore, these cells may require expansion in vitro before use as well as for future research. Their aging in vitro is thus also an important consideration. Here, we focus on bone marrow mesenchymal stem cells (BMSCs), which are unique compared to other stem cells due to their support of hematopoietic cells in addition to contributing to bone formation. BMSCs may be sensitive to age-related diseases and could perpetuate degenerative diseases in which bone remodeling is a contributory factor. Here, we review (1) the characterization of BMSCs, (2) the characterization of in vivo-aged BMSCs, (3) the characterization of in vitro-aged BMSCs, and (4) potential approaches to optimize the performance of aged BMSCs. This article is part of a Special Issue entitled "Stem Cells and Bone".
Collapse
Affiliation(s)
- Natasha Baker
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa B Boyette
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
45
|
Aguilar E, Bagó JR, Soler-Botija C, Alieva M, Rigola MA, Fuster C, Vila OF, Rubio N, Blanco J. Fast-proliferating adipose tissue mesenchymal-stromal-like cells for therapy. Stem Cells Dev 2014; 23:2908-20. [PMID: 25019281 DOI: 10.1089/scd.2014.0231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Human mesenchymal stromal cells, whether from the bone marrow or adipose tissue (hASCs), are promising cell therapy agents. However, generation of abundant cells for therapy remains to be a challenge, due to the need of lengthy expansion and the risk of accumulating genomic defects during the process. We show that hASCs can be easily induced to a reversible fast-proliferating phenotype (FP-ASCs) that allows rapid generation of a clinically useful quantity of cells in <2 weeks of culture. Expanded FP-ASCs retain their finite expansion capacity and pluripotent properties. Despite the high proliferation rate, FP-ASCs show genomic stability by array-comparative genomic hybridization, and did not generate tumors when implanted for a long time in an SCID mouse model. Comparative analysis of gene expression patterns revealed a set of genes that can be used to characterize FP-ASCs and distinguish them from hASCs. As potential candidate therapeutic agents, FP-ASCs displayed high vasculogenic capacity in Matrigel assays. Moreover, application of hASCs and FP-ASCs in a fibrin scaffold over a myocardium infarct model in SCID mice showed that both cell types can differentiate to endothelial and myocardium lineages, although FP-ASCs were more potent angiogenesis inducers than hASCs, at promoting myocardium revascularization.
Collapse
Affiliation(s)
- Elisabet Aguilar
- 1 Human DNA Variability Department, GENYO-Centre for Genomic and Oncological Research (Pfizer/University of Granada/Andalusian Regional Government) , PTS Granada, Granada, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|