1
|
Nosrati S, Gheisari M, Zare S, Dara M, Zolghadri S, Razeghian-Jahromi I. The impact of diabetic glucose concentration on viability and cardiac differentiation of mesenchymal stem cells. Tissue Cell 2024; 88:102361. [PMID: 38502970 DOI: 10.1016/j.tice.2024.102361] [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: 01/30/2024] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
INTRODUCTION Hyperglycemia may be a stumbling block for delivery of regenerative benefits of mesenchymal stem cells (MSCs) to diabetic patients with cardiovascular diseases. Our study aims to assess the viability and cardiac differentiation potential of MSCs after being exposed to diabetic glucose concentration. METHODS MSCs were extracted from rat bone marrow. Cells were characterized based on morphology, differentiation potential, and expression of mesenchymal specific markers. MTT assay was done to evaluate the viability of MSCs after treatment with different glucose concentrations. Case group was MSCs treated with diabetic concentration of glucose versus cells treated with PBS as the control group. Growth curve and population doubling time were calculated in both groups. Expression of GATA4 and troponin, as the early and late markers during cardiac differentiation, were measured following 5-azacytidine exposure. RESULTS Proliferated cells at passage three had fibroblastic-shape, was able to differentiate into adipocytes or osteocytes, and expressed CD73 and CD90. MSCs viability was gradually decreased by increasing glucose concentration. Irrespective of nicotine concentration, three-day exposure imposed more severe detrimental effects on viability compared with one-day treatment. Proliferation rate of the MSCs was lower in the case group, and they need more time for population doubling. Expression of both cardiac markers were downregulated in the case group at day three. However, their expression became higher at day seven. CONCLUSION Diabetic glucose concentration inhibits normal proliferation and cardiac differentiation of MSCs. This effect should be considered in stem cell therapy of cardiovascular patients who are concurrently affected by hyperglycemia, a common comorbidity in such individuals. Why carry out this study? What was learned from the study? FINDINGS
Collapse
Affiliation(s)
- Shadi Nosrati
- Department of Biochemistry, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Maryam Gheisari
- Department of Biochemistry, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | | |
Collapse
|
2
|
Hindle J, Williams A, Kim Y, Kim D, Patil K, Khatkar P, Osgood Q, Nelson C, Routenberg DA, Howard M, Liotta LA, Kashanchi F, Branscome H. hTERT-Immortalized Mesenchymal Stem Cell-Derived Extracellular Vesicles: Large-Scale Manufacturing, Cargo Profiling, and Functional Effects in Retinal Epithelial Cells. Cells 2024; 13:861. [PMID: 38786083 PMCID: PMC11120263 DOI: 10.3390/cells13100861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
As the economic burden associated with vision loss and ocular damage continues to rise, there is a need to explore novel treatment strategies. Extracellular vesicles (EVs) are enriched with various biological cargo, and there is abundant literature supporting the reparative and immunomodulatory properties of stem cell EVs across a broad range of pathologies. However, one area that requires further attention is the reparative effects of stem cell EVs in the context of ocular damage. Additionally, most of the literature focuses on EVs isolated from primary stem cells; the use of EVs isolated from human telomerase reverse transcriptase (hTERT)-immortalized stem cells has not been thoroughly examined. Using our large-scale EV-manufacturing platform, we reproducibly manufactured EVs from hTERT-immortalized mesenchymal stem cells (MSCs) and employed various methods to characterize and profile their associated cargo. We also utilized well-established cell-based assays to compare the effects of these EVs on both healthy and damaged retinal pigment epithelial cells. To the best of our knowledge, this is the first study to establish proof of concept for reproducible, large-scale manufacturing of hTERT-immortalized MSC EVs and to investigate their potential reparative properties against damaged retinal cells. The results from our studies confirm that hTERT-immortalized MSC EVs exert reparative effects in vitro that are similar to those observed in primary MSC EVs. Therefore, hTERT-immortalized MSCs may represent a more consistent and reproducible platform than primary MSCs for generating EVs with therapeutic potential.
Collapse
Affiliation(s)
| | - Anastasia Williams
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | - Yuriy Kim
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | | | - Kajal Patil
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | - Pooja Khatkar
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | | | - Collin Nelson
- Meso Scale Diagnostics, L.L.C., Rockville, MD 20850, USA (D.A.R.)
| | | | - Marissa Howard
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Lance A. Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 20110, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| | - Heather Branscome
- ATCC, Manassas, VA 20110, USA
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA 20110, USA (K.P.)
| |
Collapse
|
3
|
Sun W, Lv J, Guo S, Lv M. Cellular microenvironment: a key for tuning mesenchymal stem cell senescence. Front Cell Dev Biol 2023; 11:1323678. [PMID: 38111850 PMCID: PMC10725964 DOI: 10.3389/fcell.2023.1323678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) possess the ability to self-renew and differentiate into multiple cell types, making them highly suitable for use as seed cells in tissue engineering. These can be derived from various sources and have been found to play crucial roles in several physiological processes, such as tissue repair, immune regulation, and intercellular communication. However, the limited capacity for cell proliferation and the secretion of senescence-associated secreted phenotypes (SASPs) pose challenges for the clinical application of MSCs. In this review, we provide a comprehensive summary of the senescence characteristics of MSCs and examine the different features of cellular microenvironments studied thus far. Additionally, we discuss the mechanisms by which cellular microenvironments regulate the senescence process of MSCs, offering insights into preserving their functionality and enhancing their effectiveness.
Collapse
Affiliation(s)
| | | | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengzhu Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
4
|
Tian Y, Hu M, Liu X, Wang X, Lu D, Li Z, Liu Y, Zhang P, Zhou Y. ZIM1 Combined with Hydrogel Inhibits Senescence of Primary PαS Cells during In Vitro Expansion. Int J Mol Sci 2023; 24:ijms24119766. [PMID: 37298717 DOI: 10.3390/ijms24119766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Bone marrow stem cells (BMSCs) are a promising source of seed cells in bone tissue engineering, which needs a great quantity of cells. Cell senescence occurs as they are passaged, which could affect the therapeutic effects of cells. Therefore, this study aims to explore the transcriptomic differences among the uncultured and passaged cells, finding a practical target gene for anti-aging. We sorted PαS (PDGFR-α+SCA-1+CD45-TER119-) cells as BMSCs by flow cytometry analysis. The changes in cellular senescence phenotype (Counting Kit-8 (CCK-8) assay, reactive oxygen species (ROS) test, senescence-associated β-galactosidase (SA-β-Gal) activity staining, expression of aging-related genes, telomere-related changes and in vivo differentiation potential) and associated transcriptional alterations during three important cell culture processes (in vivo, first adherence in vitro, first passage, and serial passage in vitro) were studied. Overexpression plasmids of potential target genes were made and examed. Gelatin methacryloyl (GelMA) was applied to explore the anti-aging effects combined with the target gene. Aging-related genes and ROS levels increased, telomerase activity and average telomere length decreased, and SA-β-Gal activities increased as cells were passaged. RNA-seq offered that imprinted zinc-finger gene 1 (Zim1) played a critical role in anti-aging during cell culture. Further, Zim1 combined with GelMA reduced the expression of P16/P53 and ROS levels with doubled telomerase activities. Few SA-β-Gal positive cells were found in the above state. These effects are achieved at least by the activation of Wnt/β-catenin signaling through the regulation of Wnt2. The combined application of Zim1 and hydrogel could inhibit the senescence of BMSCs during in vitro expansion, which may benefit clinical application.
Collapse
Affiliation(s)
- Yueming Tian
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Menglong Hu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Xu Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Dazhuang Lu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Center for Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| |
Collapse
|
5
|
Gopalarethinam J, Nair AP, Iyer M, Vellingiri B, Subramaniam MD. Advantages of mesenchymal stem cell over the other stem cells. Acta Histochem 2023; 125:152041. [PMID: 37167794 DOI: 10.1016/j.acthis.2023.152041] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/13/2023]
Abstract
A stem cell is a particular group of cells that has the extraordinary potential to convert within the body into particular cell types. They are used to regenerate tissues and cells in the body that have been damaged or destroyed by the disease. Stem cells come in three different varieties: adult stem cells, embryonic stem cells and induced pluripotent stem cells (iPSCs). Embryonic stem cells have a high chance of immune rejection and also have ethical dilemmas and iPSCs have genetic instability. Adult stem cells are difficult to analyze and extract for research since they are frequently insufficient in native tissues. However, mesenchymal stem cells (MSC) one of the categories of adult stem cells are stromal cells with a variety of potentials that can differentiate into a wide range of cell types. MSCs can be transplanted into a variety of people without worrying about rejection because they have demonstrated the ability to prevent an adverse reaction from the immune system. These transplants have powerful anti-inflammatory and immunosuppressive effects and greatly enhance the body's inherent healing capacity. While MSCs do not offer treatment for illnesses, the idea behind them is to enable the body to recover sufficiently for a protracted reduction in symptoms. In many cases, this is sufficient to significantly enhance the patient's well-being. Inspite of several advantages some potential long-term concerns connected to MSC therapy are maldifferentiation, immunosuppression and cancerous tumor growth. In this review, we will compare the mesenchymal stem cells with other stem cells with respect to the source of origin, their properties and therapeutic applications, and discuss the MSC's disadvantages.
Collapse
Affiliation(s)
- Janani Gopalarethinam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Aswathy P Nair
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore 641021, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | - Mohana Devi Subramaniam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India.
| |
Collapse
|
6
|
Guo X, Wang J, Zou W, Wei W, Guan X, Liu J. Exploring microenvironment strategies to delay mesenchymal stem cell senescence. Stem Cells Dev 2021; 31:38-52. [PMID: 34913751 DOI: 10.1089/scd.2021.0254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as an important candidate for cell therapy and tissue regeneration. However, some limitations in translational research and therapies still exist, such as insufficient cell supply, inadequate differentiation potential, and decreased immune capacity, all of which result from replicative senescence during long-term in vitro culture. In vitro, stem cells lack a protective microenvironment owing to the absence of physical and biochemical cues compared with the in vivo niche, which provides dynamic physicochemical and biological cues. This difference results in accelerated aging after long-term in vitro culture. Therefore, it remains a great challenge to delay replicative senescence in culture. Constructing a microenvironment to delay replicative senescence of MSCs by maintaining their phenotypes, properties, and functions is a feasible strategy to solve this problem and has made measurable progress both in preclinical studies and clinical trials. Here, we review the current knowledge on the characteristics of senescent MSCs, explore the molecular mechanisms of MSCs senescence, describe the niche of MSCs, and discuss some current microenvironment strategies to delay MSCs replicative senescence that can broaden their range of therapeutic applications.
Collapse
Affiliation(s)
- Xunhui Guo
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, China;
| | - Jiayi Wang
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, Dalian, China;
| | - Wei Zou
- Liaoning Normal University, 66523, College of Life Sciences, Dalian, China;
| | - Wenjuan Wei
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Xin Guan
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Jing Liu
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| |
Collapse
|
7
|
Madry H, Grässel S, Nöth U, Relja B, Bernstein A, Docheva D, Kauther MD, Katthagen JC, Bader R, van Griensven M, Wirtz DC, Raschke MJ, Huber-Lang M. The future of basic science in orthopaedics and traumatology: Cassandra or Prometheus? Eur J Med Res 2021; 26:56. [PMID: 34127057 PMCID: PMC8200553 DOI: 10.1186/s40001-021-00521-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/20/2021] [Indexed: 12/23/2022] Open
Abstract
Orthopaedic and trauma research is a gateway to better health and mobility, reflecting the ever-increasing and complex burden of musculoskeletal diseases and injuries in Germany, Europe and worldwide. Basic science in orthopaedics and traumatology addresses the complete organism down to the molecule among an entire life of musculoskeletal mobility. Reflecting the complex and intertwined underlying mechanisms, cooperative research in this field has discovered important mechanisms on the molecular, cellular and organ levels, which subsequently led to innovative diagnostic and therapeutic strategies that reduced individual suffering as well as the burden on the society. However, research efforts are considerably threatened by economical pressures on clinicians and scientists, growing obstacles for urgently needed translational animal research, and insufficient funding. Although sophisticated science is feasible and realized in ever more individual research groups, a main goal of the multidisciplinary members of the Basic Science Section of the German Society for Orthopaedics and Trauma Surgery is to generate overarching structures and networks to answer to the growing clinical needs. The future of basic science in orthopaedics and traumatology can only be managed by an even more intensified exchange between basic scientists and clinicians while fuelling enthusiasm of talented junior scientists and clinicians. Prioritized future projects will master a broad range of opportunities from artificial intelligence, gene- and nano-technologies to large-scale, multi-centre clinical studies. Like Prometheus in the ancient Greek myth, transferring the elucidating knowledge from basic science to the real (clinical) world will reduce the individual suffering from orthopaedic diseases and trauma as well as their socio-economic impact.
Collapse
Affiliation(s)
- Henning Madry
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg, Germany
| | - Susanne Grässel
- Experimental Orthopedics, Department of Orthopedic Surgery, University of Regensburg, Regensburg, Germany
| | - Ulrich Nöth
- Department of Orthopaedics and Trauma Surgery, Evangelisches Waldkrankenhaus Berlin Spandau, Berlin, Germany
| | - Borna Relja
- Experimental Radiology, University Clinic for Radiology and Nuclear Medicine, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Anke Bernstein
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Max Daniel Kauther
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Essen, Essen, Germany
| | - Jan Christoph Katthagen
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Rainer Bader
- Department of Orthopaedics, Research Lab for Biomechanics and Implant Technology, Rostock University Medical Center, Rostock, Germany
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN-Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dieter C Wirtz
- Department of Orthopaedics and Trauma Surgery, University Hopsital Bonn, Bonn, Germany
| | - Michael J Raschke
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Helmholzstr. 8/1, Ulm, Germany.
| |
Collapse
|
8
|
Vasanthan J, Gurusamy N, Rajasingh S, Sigamani V, Kirankumar S, Thomas EL, Rajasingh J. Role of Human Mesenchymal Stem Cells in Regenerative Therapy. Cells 2020; 10:E54. [PMID: 33396426 PMCID: PMC7823630 DOI: 10.3390/cells10010054] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells which can proliferate and replace dead cells in the body. MSCs also secrete immunomodulatory molecules, creating a regenerative microenvironment that has an excellent potential for tissue regeneration. MSCs can be easily isolated and grown in vitro for various applications. For the past two decades, MSCs have been used in research, and many assays and tests have been developed proving that MSCs are an excellent cell source for therapy. This review focusses on quality control parameters required for applications of MSCs including colony formation, surface markers, differentiation potentials, and telomere length. Further, the specific mechanisms of action of MSCs under various conditions such as trans-differentiation, cell fusion, mitochondrial transfer, and secretion of extracellular vesicles are discussed. This review aims to underline the applications and benefits of MSCs in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Edwin L. Thomas
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
9
|
Changes in Stemness Properties, Differentiation Potential, Oxidative Stress, Senescence and Mitochondrial Function in Wharton's Jelly Stem Cells of Umbilical Cords of Mothers with Gestational Diabetes Mellitus. Stem Cell Rev Rep 2020; 15:415-426. [PMID: 30645713 DOI: 10.1007/s12015-019-9872-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gestational diabetes mellitus (GDM) has been associated with an increased risk of maternal and neonatal morbidity. The Wharton's jelly (WJ) of the umbilical cord (UC) is a useful indicator of the deleterious effects of hyperglycemia on fetal tissues as it represents the fetus embryologically, physiologically and genetically. We studied WJ mesenchymal stem cells (hWJSCs) from UC from mothers without GDM (Normal; n = 3); insulin-controlled GDM mothers (GDMi; n = 3) and diet-controlled GDM mothers (GDMd; n = 3)]. Cell proliferation, stemness markers, telomerase, osteogenic and chondrogenic differentiation, antioxidant enzymes and gene expression for mitochondrial function (ND2, TFAM, PGC1α, and NDUFB9) were significantly lower in GDMi-hWJSCs and GDMd-hWJSCs compared to normal hWJSCs (P < 0.05). On the other hand, cell cycle inhibitors (p16, p21, p27) and p53 were remarkably up-regulated in GDMi-hWJSCs and GDMd-hWJSCs compared to normal hWJSCs. The results from this study confirmed that maternal hyperglycemia even though managed with insulin or diet, induced changes in the properties of the WJ and its cells. These changes may also be observed in fetal tissues and if true, prevention of the onset of gestational diabetes should be a priority over management. Generation of tissues that simulate those of the fetus such as pancreatic and cardiovascular cells from GDM-hWJSCs by direct differentiation or via induced pluripotent stem cell reprogramming provide possible platforms to evaluate the effects of glucose on specific fetal organ.
Collapse
|
10
|
Lavrentieva A, Hoffmann A, Lee-Thedieck C. Limited Potential or Unfavorable Manipulations? Strategies Toward Efficient Mesenchymal Stem/Stromal Cell Applications. Front Cell Dev Biol 2020; 8:316. [PMID: 32509777 PMCID: PMC7248306 DOI: 10.3389/fcell.2020.00316] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Despite almost 50 years of research and over 20 years of preclinical and clinical studies, the question of curative potential of mesenchymal stem/stromal cells (MSCs) is still widely discussed in the scientific community. Non-reproducible treatment outcomes or even absence of treatment effects in comparison to control groups challenges the potential of these cells for routine application both in tissue engineering and in regenerative medicine. One of the reasons of such outcomes is non-standardized and often disadvantageous ex vivo manipulation of MSCs prior therapy. In most cases, clinically relevant cell numbers for MSC-based therapies can be only obtained by in vitro expansion of isolated cells. In this mini review, we will discuss point by point possible pitfalls in the production of human MSCs for cell therapies, without consideration of material-based applications. Starting with cell source, choice of donor and recipient, as well as isolation methods, we will then discuss existing expansion protocols (two-/three-dimensional cultivation, basal medium, medium supplements, static/dynamic conditions, and hypoxic/normoxic conditions) and influence of these strategies on the cell functionality after implantation. The role of potency assays will also be addressed. The final aim of this mini review is to illustrate the heterogeneity of current strategies for gaining MSCs for clinical applications with their strengths and weaknesses. Only a careful consideration and standardization of all pretreatment processes/methods for the different applications of MSCs will ensure robust and reproducible performance of these cell populations in the different experimental and clinical settings.
Collapse
Affiliation(s)
| | - Andrea Hoffmann
- Department of Orthopaedic Surgery, Graded Implants and Regenerative Strategies, Hannover Medical School, Hanover, Germany
| | - Cornelia Lee-Thedieck
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hanover, Germany
| |
Collapse
|
11
|
Xu C, Xie N, Su Y, Sun Z, Liang Y, Zhang N, Liu D, Jia S, Xing X, Han L, Li G, Tong T, Chen J. HnRNP F/H associate with hTERC and telomerase holoenzyme to modulate telomerase function and promote cell proliferation. Cell Death Differ 2019; 27:1998-2013. [PMID: 31863069 PMCID: PMC7244589 DOI: 10.1038/s41418-019-0483-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
Human telomerase RNA component hTERC comprises multiple motifs that contribute to hTERC biogenesis, holoenzyme activity, and enzyme recruitment to telomeres. hTERC contains several guanine tracts (G-tracts) at its 5′-end, but its associated proteins and potential roles in telomerase function are still poorly understood. The heterogeneous nuclear ribonucleoproteins F, H1, and H2 (hnRNP F/H) are splicing factors that preferentially bind to poly(G)-rich sequences RNA. Here, we demonstrate that hnRNP F/H associate with both hTERC and telomerase holoenzyme to regulate telomerase activity. We reveal hnRNP F/H bind to the 5′-end region of hTERC in vitro and in vivo, and identify the first three G-tracts of hTERC and qRRM1 domain of hnRNP F/H are required for their interaction. Furthermore, hnRNP F/H also directly interact with telomerase holoenzyme. Functionally, we show that hnRNP F/H plays important roles in modulating telomerase activity and telomere length. Moreover, hnRNP F/H deletion greatly impair cancer and stem cell proliferation, and induce stem cell senescence, while hnRNP F/H overexpression delay stem cell senescence. Collectively, our findings unveil a novel role of hnRNP F/H as the binding partners of hTERC and telomerase holoenzyme to regulate telomerase function.
Collapse
Affiliation(s)
- Chenzhong Xu
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Nan Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Yuanyuan Su
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Zhaomeng Sun
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Yao Liang
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Na Zhang
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Doudou Liu
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Shuqin Jia
- Department of Molecular Diagnostics, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaofang Xing
- Department of Molecular Diagnostics, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Limin Han
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Guodong Li
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China.
| |
Collapse
|
12
|
Karaöz E, Tepeköy F. Differentiation Potential and Tumorigenic Risk of Rat Bone Marrow Stem Cells Are Affected By Long-Term In Vitro Expansion. Turk J Haematol 2019; 36:255-265. [PMID: 31284704 PMCID: PMC6863016 DOI: 10.4274/tjh.galenos.2019.2019.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective: Mesenchymal stem cells (MSCs) have the capacity for extensive expansion and adipogenic, osteogenic, chondrogenic, myogenic, and neural differentiation in vitro. The aim of our study was to determine stemness, differentiation potential, telomerase activity, and ultrastructural characteristics of long-term cultured rat bone marrow (rBM)-MSCs. Materials and Methods: rBM-MSCs from passages 3, 50, and 100 (P3, P50, and P100) were evaluated through immunocytochemistry, reverse transcription-polymerase chain reaction, telomerase activity assays, and electron microscopy. Results: A dramatic reduction in the levels of myogenic markers actin and myogenin was detected in P100. Osteogenic markers Coll1, osteonectin (Sparc), and osteocalcin as well as neural marker c-Fos and chondrogenic marker Coll2 were significantly reduced in P100 compared to P3 and P50. Osteogenic marker bone morphogenic protein-2 (BMP2) and adipogenic marker peroxisome proliferator-activated receptor gamma (Pparγ) expression was reduced in late passages. The expression of stemness factor Rex-1 was lower in P100, whereas Oct4 expression was decreased in P50 compared to P3 and P100. Increased telomerase activity was observed in long-term cultured cells, signifying tumorigenic risk. Electron microscopic evaluations revealed ultrastructural changes such as smaller number of organelles and increased amount of autophagic vacuoles in the cytoplasm in long-term cultured rBM-MSCs. Conclusion: This study suggests that long-term culture of rBM-MSCs leads to changes in differentiation potential and increased tumorigenic risk.
Collapse
Affiliation(s)
- Erdal Karaöz
- İstinye University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Turkey,İstinye University Center for Stem Cell and Tissue Engineering Research and Practice, İstanbul, Turkey,Center for Regenerative Medicine and Stem Cell Research and Manufacturing (LivMedCell), İstanbul, Turkey
| | - Filiz Tepeköy
- İstinye University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Turkey,Altınbaş University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Turkey
| |
Collapse
|
13
|
Neri S. Genetic Stability of Mesenchymal Stromal Cells for Regenerative Medicine Applications: A Fundamental Biosafety Aspect. Int J Mol Sci 2019; 20:ijms20102406. [PMID: 31096604 PMCID: PMC6566307 DOI: 10.3390/ijms20102406] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) show widespread application for a variety of clinical conditions; therefore, their use necessitates continuous monitoring of their safety. The risk assessment of mesenchymal stem cell-based therapies cannot be separated from an accurate and deep knowledge of their biological properties and in vitro and in vivo behavior. One of the most relevant safety issues is represented by the genetic stability of MSCs, that can be altered during in vitro manipulation, frequently required before clinical application. MSC genetic stability has the potential to influence the transformation and the therapeutic effect of these cells. At present, karyotype evaluation represents the definitely prevailing assessment of MSC stability, but DNA alterations of smaller size should not be underestimated. This review will focus on current scientific knowledge about the genetic stability of mesenchymal stem cells. The techniques used and possible improvements together with regulatory aspects will also be discussed.
Collapse
Affiliation(s)
- Simona Neri
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
14
|
Antebi B, Walker KP, Mohammadipoor A, Rodriguez LA, Moore RK, Cancio LC, Batchinsky AI. Bench-to-bedside optimization of mesenchymal stem cell isolation, processing, and expansion for in vivo administration. Regen Med 2019; 14:279-293. [PMID: 31070521 DOI: 10.2217/rme-2018-0043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: In this study, we aimed at identifying the optimal conditions for isolation, processing and expansion of mesenchymal stem cells (MSCs). Methods: Porcine bone marrow was obtained from either small- or large-volume bone marrow aspirate (BMA). Next, three BMA processing methods were compared. Finally, the best condition was selected from various culture parameters, including basal media, supplementation and seeding density. Results: Our results demonstrate that a small-volume BMA and direct plating yields significantly higher concentration of MSCs. Basal media supplementation with 10% platelet lysate and seeding density of 1000 cells/cm2 can generate large numbers of multipotent MSCs with augmented function and low population doublings. Conclusion: This work provides guidance for preparation of robust MSCs for future clinical trials.
Collapse
Affiliation(s)
- Ben Antebi
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Kerfoot P Walker
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.,Oak Ridge Institute for Science & Education, Oak Ridge, TN 37831-0117, USA
| | - Arezoo Mohammadipoor
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.,Oak Ridge Institute for Science & Education, Oak Ridge, TN 37831-0117, USA
| | - Luis A Rodriguez
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Robbie K Moore
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Leopoldo C Cancio
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Andriy I Batchinsky
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.,The Geneva Foundation, Tacoma, WA 98402, USA
| |
Collapse
|
15
|
Alteration of Apoptosis during Differentiation in Human Dental Pulp-Derived Mesenchymal Stem Cell. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2019. [DOI: 10.12750/jarb.34.1.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
16
|
López-Alcorocho JM, Guillén-Vicente I, Rodríguez-Iñigo E, Guillén-Vicente M, Fernández-Jaén TF, Caballero R, Casqueiro M, Najarro P, Abelow S, Guillén-García P. Study of Telomere Length in Preimplanted Cultured Chondrocytes. Cartilage 2019; 10:36-42. [PMID: 29322876 PMCID: PMC6376562 DOI: 10.1177/1947603517749918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
DESIGN In the process of cell division, the extremes of the eukaryotic chromosomes are progressively shortening, and this phenomenon is related to cell degeneration and senescence. The treatment of cartilage lesions with autologous chondrocytes implies that cells proliferate in an artificial environment. We have studied the viability of cultured chondrocytes after measurement of their telomere length before implantation. METHODS Articular cartilage biopsies (B1, B2, and B3) were obtained from 3 patients (2 males and 1 female) with knee cartilage defects, who were going to be treated with chondrocyte implantation. Chondrocytes were cultured in DMEM with autologous serum. After the third passage, an aliquot of 1 million cells was removed to estimate the telomere length and the remaining cells were implanted. Telomere length was measured by quantitative fluorescent in situ hybridization (Q-FISH). Patients' clinical outcome was determined preoperatively, and 12 and 24 months postimplantation with the International Knee Documentation Committee (IKDC) questionnaire. RESULTS After chondrocyte implantation, IKDC score doubled at 12 and 24 months with regard to the basal value. After 3 passages, chondrocytes were cultured for a mean of 45.67 days, the mean duplication time being 4.53 days and the mean number of cell divisions being 10.04 during the culture period. The 20th percentile of telomere lengths were 6.84, 6.96, and 7.06 kbp and the median telomere lengths 10.30, 10.47, and 10.73 kbp, respectively. No significant correlation was found between IKDC score and telomere length. CONCLUSION Culturing autologous chondrocytes for implantation is not related to cell senescence in terms of telomere length.
Collapse
Affiliation(s)
- Juan Manuel López-Alcorocho
- Research Unit, Clínica Cemtro, Madrid, Spain,Juan Manuel López-Alcorocho, Research Unit, Clínica Cemtro, C/ Ventisquero de la Condesa, 42, 28035 Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W, Osowski A, Wojtkiewicz J. Articular Cartilage Aging-Potential Regenerative Capacities of Cell Manipulation and Stem Cell Therapy. Int J Mol Sci 2018; 19:E623. [PMID: 29470431 PMCID: PMC5855845 DOI: 10.3390/ijms19020623] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/11/2018] [Accepted: 02/16/2018] [Indexed: 12/13/2022] Open
Abstract
Changes in articular cartilage during the aging process are a stage of natural changes in the human body. Old age is the major risk factor for osteoarthritis but the disease does not have to be an inevitable consequence of aging. Chondrocytes are particularly prone to developing age-related changes. Changes in articular cartilage that take place in the course of aging include the acquisition of the senescence-associated secretory phenotype by chondrocytes, a decrease in the sensitivity of chondrocytes to growth factors, a destructive effect of chronic production of reactive oxygen species and the accumulation of the glycation end products. All of these factors affect the mechanical properties of articular cartilage. A better understanding of the underlying mechanisms in the process of articular cartilage aging may help to create new therapies aimed at slowing or inhibiting age-related modifications of articular cartilage. This paper presents the causes and consequences of cellular aging of chondrocytes and the biological therapeutic outlook for the regeneration of age-related changes of articular cartilage.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| |
Collapse
|
18
|
Samsonraj RM, Raghunath M, Nurcombe V, Hui JH, van Wijnen AJ, Cool SM. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem Cells Transl Med 2017; 6:2173-2185. [PMID: 29076267 PMCID: PMC5702523 DOI: 10.1002/sctm.17-0129] [Citation(s) in RCA: 471] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC) hold great potential for regenerative medicine because of their ability for self-renewal and differentiation into tissue-specific cells such as osteoblasts, chondrocytes, and adipocytes. MSCs orchestrate tissue development, maintenance and repair, and are useful for musculoskeletal regenerative therapies to treat age-related orthopedic degenerative diseases and other clinical conditions. Importantly, MSCs produce secretory factors that play critical roles in tissue repair that support both engraftment and trophic functions (autocrine and paracrine). The development of uniform protocols for both preparation and characterization of MSCs, including standardized functional assays for evaluation of their biological potential, are critical factors contributing to their clinical utility. Quality control and release criteria for MSCs should include cell surface markers, differentiation potential, and other essential cell parameters. For example, cell surface marker profiles (surfactome), bone-forming capacities in ectopic and orthotopic models, as well as cell size and granularity, telomere length, senescence status, trophic factor secretion (secretome), and immunomodulation, should be thoroughly assessed to predict MSC utility for regenerative medicine. We propose that these and other functionalities of MSCs should be characterized prior to use in clinical applications as part of comprehensive and uniform guidelines and release criteria for their clinical-grade production to achieve predictably favorable treatment outcomes for stem cell therapy. Stem Cells Translational Medicine 2017;6:2173-2185.
Collapse
Affiliation(s)
- Rebekah M. Samsonraj
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore
- Department of Orthopaedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Michael Raghunath
- Department of Biomedical EngineeringNational University of SingaporeSingapore
- Center for Cell Biology and Tissue Engineering, Competence Center for Tissue Engineering and Substance Testing (TEDD)Institute for Chemistry and Biotechnology, ZHAW School of Life Sciences and Facility Management, Zurich University of Applied SciencesSwitzerland
| | - Victor Nurcombe
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
| | - James H. Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | | | - Simon M. Cool
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| |
Collapse
|
19
|
Szychlinska MA, Stoddart MJ, D'Amora U, Ambrosio L, Alini M, Musumeci G. Mesenchymal Stem Cell-Based Cartilage Regeneration Approach and Cell Senescence: Can We Manipulate Cell Aging and Function? TISSUE ENGINEERING PART B-REVIEWS 2017; 23:529-539. [PMID: 28514935 DOI: 10.1089/ten.teb.2017.0083] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aging is the most prominent risk factor triggering several degenerative diseases, such as osteoarthritis (OA). Due to its poor self-healing capacity, once injured cartilage needs to be reestablished. This process might be approached through resorting to cell-based therapies and/or tissue engineering. Human mesenchymal stem cells (MSCs) represent a promising approach due to their chondrogenic differentiation potential. Presently, in vitro chondrogenic differentiation of MSCs is limited by two main reasons as follows: aging of MSCs, which determines the loss of cell proliferative and differentiation capacity and MSC-derived chondrocyte hypertrophic differentiation, which limits the use of these cells in cartilage tissue regeneration approach. The effect of aging on MSCs is fundamental for stem cell-based therapy development, especially in older subjects. In the present review we focus on homeostasis alterations occurring in MSC-derived chondrocytes during in vitro aging. Moreover, we deal with potential cell aging regulation approaches, such as cell stimulation through telomerase activators, mechanical strain, and epigenetic regulation. Future investigations in this field might provide new insights into innovative strategies for cartilage regeneration and potentially inspire novel therapeutic approaches for OA treatment.
Collapse
Affiliation(s)
- Marta A Szychlinska
- 1 Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Catania, Italy
| | - Martin J Stoddart
- 2 Musculoskeletal Regeneration, AO Research Institute Davos , Davos Platz, Switzerland
| | - Ugo D'Amora
- 3 Institute of Polymers , Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Luigi Ambrosio
- 3 Institute of Polymers , Composites and Biomaterials, National Research Council of Italy, Naples, Italy .,4 Department of Chemical Science and Materials Technology, National Research Council of Italy , Rome, Italy
| | - Mauro Alini
- 2 Musculoskeletal Regeneration, AO Research Institute Davos , Davos Platz, Switzerland
| | - Giuseppe Musumeci
- 1 Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Catania, Italy .,5 Department of Health, Institut des Etudes Universitaries , UniPoliSI, Veyras, Switzerland
| |
Collapse
|
20
|
Zhang D, Lu H, Chen Z, Wang Y, Lin J, Xu S, Zhang C, Wang B, Yuan Z, Feng X, Jiang X, Pan J. High glucose induces the aging of mesenchymal stem cells via Akt/mTOR signaling. Mol Med Rep 2017; 16:1685-1690. [PMID: 28656269 PMCID: PMC5562095 DOI: 10.3892/mmr.2017.6832] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 02/23/2017] [Indexed: 02/07/2023] Open
Abstract
It has previously been demonstrated that glucose is important in the process of stem cell aging. However, the mechanisms of cell senescence induced by high glucose (HG) remain to be elucidated. The preliminary study indicated that D-galactose induced mesenchymal stem cell (MSCs) aging. The present study demonstrated, following treatment with 11.0 or 22.0 mM HG for 14 days, that HG significantly promoted MSCs aging and the expression levels of phosphorylated (p-)phosphatidylinositol 3-kinase/protein kinase B (Akt) and p-mammalian target of rapamycin signaling (mTOR) in the HG groups were increased compared with the control group. However, following Akt inhibition with 1.0 or 10.0 nM MK-2206, which is an Akt-specific small molecule inhibitor, the senescence-cell value in the HG group was significantly decreased compared with the control group. These results indicated that HG induced MSCs senescence and this effect was primarily mediated via the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Dayong Zhang
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Huifei Lu
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Zhongxing Chen
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Yayan Wang
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Jiuzhou Lin
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Shan Xu
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Chong Zhang
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Baoming Wang
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Zhanggen Yuan
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| | - Xiao Feng
- Clinical Laboratory, The Second People's Hospital of Hangzhou, Hangzhou, Zhejiang 310015, P.R. China
| | - Xuefan Jiang
- Department of Otorhinolaryngology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jianping Pan
- Department of Clinical Medicine, Zhejiang University City College School of Medicine, Hangzhou, Zhejiang 310015, P.R. China
| |
Collapse
|
21
|
Li Y, Wu Q, Wang Y, Li L, Bu H, Bao J. Senescence of mesenchymal stem cells (Review). Int J Mol Med 2017; 39:775-782. [PMID: 28290609 DOI: 10.3892/ijmm.2017.2912] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 01/13/2017] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in cell-based therapy for various diseases, due to their immunomodulatory and inflammatory effects. However, the function of MSCs is known to decline with age, a process that is called senescence. To date, the process of MSC senescence remains unknown as in-depth understanding of the mechanisms involved in cellular senescence is lacking. First, senescent MSCs are so heterogeneous that not all of them express the same phenotypic markers. In addition, the genes and signaling pathways which regulate this process in MSCs are still unknown. Thus, an understanding of the molecular processes controlling MSC senescence is crucial to determining the drivers and effectors of age-associated MSC dysfunction. Moreover, the proper use of MSCs for clinical application requires a general understanding of the MSC aging process. Furthermore, such knowledge is essential for the development of therapeutic interventions that can slow or reverse age-related degenerative changes to enhance repair processes and maintain healthy function in aging tissues. To further clarify the properties of senescent cells, as well as to present significant findings from studies on the mechanisms of cellular aging, we summarize these biological features in the senescence of MSCs in this scenario. This review summarizes recent advances in our understanding of the markers and differentiation potential indicating MSC senescence, as well as factors affecting MSC senescence with particular emphasis on the roles of oxidative stress, intrinsic changes in telomere shortening, histone deacetylase and DNA methyltransferase, genes and signaling pathways and immunological properties.
Collapse
Affiliation(s)
- Yi Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qiong Wu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yujia Wang
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong Bu
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ji Bao
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
22
|
Challenges for Cartilage Regeneration. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2017. [DOI: 10.1007/978-3-662-53574-5_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Stab BR, Martinez L, Grismaldo A, Lerma A, Gutiérrez ML, Barrera LA, Sutachan JJ, Albarracín SL. Mitochondrial Functional Changes Characterization in Young and Senescent Human Adipose Derived MSCs. Front Aging Neurosci 2016; 8:299. [PMID: 28018212 PMCID: PMC5156959 DOI: 10.3389/fnagi.2016.00299] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are highly dynamic organelles that in response to the cell's bio-energetic state continuously undergo structural remodeling fission and fusion processes. This mitochondrial dynamic activity has been implicated in cell cycle, autophagy, and age-related diseases. Adult tissue-derived mesenchymal stromal/stem cells present a therapeutic potential. However, to obtain an adequate mesenchymal stromal/stem cell number for clinical use, extensive in vitro expansion is required. Unfortunately, these cells undergo replicative senescence rapidly by mechanisms that are not well understood. Senescence has been associated with metabolic changes in the oxidative state of the cell, a process that has been also linked to mitochondrial fission and fusion events, suggesting an association between mitochondrial dynamics and senescence. In the present work, we studied the mitochondrial structural remodeling process of mesenchymal stromal/stem cells isolated from adipose tissue in vitro to determine if mitochondrial phenotypic changes were associated with mesenchymal stromal/stem cell senescence. For this purpose, mitochondrial dynamics and oxidative state of stromal/stem cell were compared between young and old cells. With increased cell passage, we observed a significant change in cell morphology that was associated with an increase in β-galactosidase activity. In addition, old cells (population doubling seven) also showed increased mitochondrial mass, augmented superoxide production, and decreased mitochondrial membrane potential. These changes in morphology were related to slightly levels increases in mitochondrial fusion proteins, Mitofusion 1 (MFN1), and Dynamin-related GTPase (OPA1). Collectively, our results showed that adipose tissue-derived MSCs at population doubling seven developed a senescent phenotype that was characterized by metabolic cell changes that can lead to mitochondrial fusion.
Collapse
Affiliation(s)
- Bernd R Stab
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Laura Martinez
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Adriana Grismaldo
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Alejandra Lerma
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - María L Gutiérrez
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad JaverianaBogotá, Colombia; Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad JaverianaBogotá, Colombia
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Jhon J Sutachan
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Sonia L Albarracín
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| |
Collapse
|
24
|
Toh WS, Brittberg M, Farr J, Foldager CB, Gomoll AH, Hui JHP, Richardson JB, Roberts S, Spector M. Cellular senescence in aging and osteoarthritis. Acta Orthop 2016; 87:6-14. [PMID: 27658487 PMCID: PMC5389431 DOI: 10.1080/17453674.2016.1235087] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
- It is well accepted that age is an important contributing factor to poor cartilage repair following injury, and to the development of osteoarthritis. Cellular senescence, the loss of the ability of cells to divide, has been noted as the major factor contributing to age-related changes in cartilage homeostasis, function, and response to injury. The underlying mechanisms of cellular senescence, while not fully understood, have been associated with telomere erosion, DNA damage, oxidative stress, and inflammation. In this review, we discuss the causes and consequences of cellular senescence, and the associated biological challenges in cartilage repair. In addition, we present novel strategies for modulation of cellular senescence that may help to improve cartilage regeneration in an aging population.
Collapse
Affiliation(s)
- Wei Seong Toh
- Faculty of Dentistry, National University of Singapore,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore,Correspondence:
| | - Mats Brittberg
- Cartilage Research Unit, University of Gothenburg, Gothenburg,Department of Orthopaedics, Kungsbacka Hospital, Kungsbacka, Sweden
| | - Jack Farr
- Indiana University School of Medicine, OrthoIndy Cartilage Restoration Center, Indianapolis, IN, USA
| | | | - Andreas H Gomoll
- Cartilage Repair Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - James Hoi Po Hui
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore,Cartilage Repair Program, Therapeutic Tissue Engineering Laboratory, Department of Orthopaedic Surgery, National University Health System, National University of Singapore, Singapore
| | - James B Richardson
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire,Institute for Science andTechnology in Medicine, Keele University, Keele, Staffordshire, UK
| | - Sally Roberts
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire,Institute for Science andTechnology in Medicine, Keele University, Keele, Staffordshire, UK
| | - Myron Spector
- Department of Orthopaedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA,Tissue Engineering Laboratories, VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
25
|
Mennan C, Brown S, McCarthy H, Mavrogonatou E, Kletsas D, Garcia J, Balain B, Richardson J, Roberts S. Mesenchymal stromal cells derived from whole human umbilical cord exhibit similar properties to those derived from Wharton's jelly and bone marrow. FEBS Open Bio 2016; 6:1054-1066. [PMID: 27833846 PMCID: PMC5095143 DOI: 10.1002/2211-5463.12104] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 05/27/2016] [Accepted: 07/20/2016] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSC) can be isolated from several regions of human umbilical cords, including Wharton's jelly (WJ), artery, vein or cord lining. These MSC appear to be immune privileged and are promising candidates for cell therapy. However, isolating MSC from WJ, artery, vein or cord lining requires time-consuming tissue dissection. MSC can be obtained easily via briefly digesting complete segments of the umbilical cord, likely containing heterogenous or mixed populations of MSC (MC-MSC). MC-MSC are generally less well characterized than WJ-MSC, but nevertheless represent a potentially valuable population of MSC. This study aimed to further characterize MC-MSC in comparison to WJ-MSC and also the better-characterized bone marrow-derived MSC (BM-MSC). MC-MSC proliferated faster, with significantly faster doubling times reaching passage one 8.8 days sooner and surviving longer in culture than WJ-MSC. All MSC retained the safety aspect of reducing telomere length with increasing passage number. MSC were also assessed for their ability to suppress T-cell proliferation and for the production of key markers of pluripotency, embryonic stem cells, tolerogenicity (CD40, CD80, CD86 and HLA-DR) and immunomodulation (indoleamine 2,3-dioxygenase [IDO] and HLA-G). The MC-MSC population displayed all of the positive attributes of WJ-MSC and BM-MSC, but they were more efficient to obtain and underwent more population doublings than from WJ, suggesting that MC-MSC are promising candidates for allogeneic cell therapy in regenerative medicine.
Collapse
Affiliation(s)
- Claire Mennan
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| | - Sharon Brown
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| | - Helen McCarthy
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| | - Eleni Mavrogonatou
- Laboratory of Cell Proliferation and AgeingInstitute of Biosciences and ApplicationsNational Centre for Scientific Research “Demokritos”AthensGreece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and AgeingInstitute of Biosciences and ApplicationsNational Centre for Scientific Research “Demokritos”AthensGreece
| | - John Garcia
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| | - Birender Balain
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| | - James Richardson
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| | - Sally Roberts
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust & Institute of Science & Technology in MedicineKeele UniversityOswestryShropshireUK
| |
Collapse
|
26
|
Cassar L, Nicholls C, Pinto AR, Chen R, Wang L, Li H, Liu JP. TGF-beta receptor mediated telomerase inhibition, telomere shortening and breast cancer cell senescence. Protein Cell 2016; 8:39-54. [PMID: 27696331 PMCID: PMC5233610 DOI: 10.1007/s13238-016-0322-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 08/03/2016] [Indexed: 01/09/2023] Open
Abstract
Human telomerase reverse transcriptase (hTERT) plays a central role in telomere lengthening for continuous cell proliferation, but it remains unclear how extracellular cues regulate telomerase lengthening of telomeres. Here we report that the cytokine bone morphogenetic protein-7 (BMP7) induces the hTERT gene repression in a BMPRII receptor- and Smad3-dependent manner in human breast cancer cells. Chonic exposure of human breast cancer cells to BMP7 results in short telomeres, cell senescence and apoptosis. Mutation of the BMPRII receptor, but not TGFbRII, ACTRIIA or ACTRIIB receptor, inhibits BMP7-induced repression of the hTERT gene promoter activity, leading to increased telomerase activity, lengthened telomeres and continued cell proliferation. Expression of hTERT prevents BMP7-induced breast cancer cell senescence and apoptosis. Thus, our data suggest that BMP7 induces breast cancer cell aging by a mechanism involving BMPRII receptor- and Smad3-mediated repression of the hTERT gene.
Collapse
Affiliation(s)
- Lucy Cassar
- Molecular Signaling Laboratory, Department of Immunology, Central Eastern Clinical School, Monash University, Prahran, VIC, 3181, Australia
| | - Craig Nicholls
- Molecular Signaling Laboratory, Department of Immunology, Central Eastern Clinical School, Monash University, Prahran, VIC, 3181, Australia
| | - Alex R Pinto
- Molecular Signaling Laboratory, Department of Immunology, Central Eastern Clinical School, Monash University, Prahran, VIC, 3181, Australia
| | - Ruping Chen
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, 311121, Zhejiang Province, China
| | - Lihui Wang
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, 311121, Zhejiang Province, China
| | - He Li
- Molecular Signaling Laboratory, Department of Immunology, Central Eastern Clinical School, Monash University, Prahran, VIC, 3181, Australia
| | - Jun-Ping Liu
- Molecular Signaling Laboratory, Department of Immunology, Central Eastern Clinical School, Monash University, Prahran, VIC, 3181, Australia. .,Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, 311121, Zhejiang Province, China.
| |
Collapse
|
27
|
El Atat O, Antonios D, Hilal G, Hokayem N, Abou-Ghoch J, Hashim H, Serhal R, Hebbo C, Moussa M, Alaaeddine N. An Evaluation of the Stemness, Paracrine, and Tumorigenic Characteristics of Highly Expanded, Minimally Passaged Adipose-Derived Stem Cells. PLoS One 2016; 11:e0162332. [PMID: 27632538 PMCID: PMC5024991 DOI: 10.1371/journal.pone.0162332] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/22/2016] [Indexed: 12/13/2022] Open
Abstract
The use of adipose-derived stem cells (ADSC) in regenerative medicine is rising due to their plasticity, capacity of differentiation and paracrine and trophic effects. Despite the large number of cells obtained from adipose tissue, it is usually not enough for therapeutic purposes for many diseases or cosmetic procedures. Thus, there is the need for culturing and expanding cells in-vitro for several weeks remain. Our aim is to investigate if long- term proliferation with minimal passaging will affect the stemness, paracrine secretions and carcinogenesis markers of ADSC. The immunophenotypic properties and aldehyde dehydrogenase (ALDH) activity of the initial stromal vascular fraction (SVF) and serially passaged ADSC were observed by flow cytometry. In parallel, the telomerase activity and the relative expression of oncogenes and tumor suppressor genes were assessed by q-PCR. We also assessed the cytokine secretion profile of passaged ADSC by an ELISA. The expanded ADSC retain their morphological and phenotypical characteristics. These cells maintained in culture for up to 12 weeks until P4, possessed stable telomerase and ALDH activity, without having a TP53 mutation. Furthermore, the relative expression levels of TP53, RB, and MDM2 were not affected while the relative expression of c-Myc decreased significantly. Finally, the levels of the secretions of PGE2, STC1, and TIMP2 were not affected but the levels of IL-6, VEGF, and TIMP 1 significantly decreased at P2. Our results suggest that the expansion of passaged ADSC does not affect the differentiation capacity of stem cells and does not confer a cancerous state or capacity in vitro to the cells.
Collapse
Affiliation(s)
- Oula El Atat
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Diane Antonios
- Toxicology Laboratory, Faculty of Pharmacy, St. Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Nabil Hokayem
- Department of Plastic& Reconstructive Surgery, Hotel Dieu de France, and Faculty of Medicine St Joseph University, Beirut, Lebanon
| | - Joelle Abou-Ghoch
- Medical Genetics Unit, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Hussein Hashim
- Department of Plastic& Reconstructive Surgery, Fuad Khoury Hospital, Beirut, Lebanon
| | - Rim Serhal
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Clara Hebbo
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Mayssam Moussa
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
| | - Nada Alaaeddine
- Regenerative Medicine and Inflammation Laboratory, Faculty of Medicine, St. Joseph University, Beirut, Lebanon
- * E-mail:
| |
Collapse
|
28
|
Pizzute T, Zhang Y, He F, Pei M. Ascorbate-dependent impact on cell-derived matrix in modulation of stiffness and rejuvenation of infrapatellar fat derived stem cells toward chondrogenesis. Biomed Mater 2016; 11:045009. [PMID: 27508528 PMCID: PMC5004760 DOI: 10.1088/1748-6041/11/4/045009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Developing an in vitro microenvironment using cell-derived decellularized extracellular matrix (dECM) is a promising approach to efficiently expand adult stem cells for cartilage engineering and regeneration. Ascorbic acid serves as a critical stimulus for cells to synthesize collagens, which constitute the major component of dECM. In this study, we hypothesized that optimization of ascorbate treatment would maximize the rejuvenation effect of dECM on expanded stem cells from human infrapatellar fat pad in both proliferation and chondrogenic differentiation. In the duration regimen study, we found that dECM without L-ascorbic acid phosphate (AA) treatment, exhibiting lower stiffness measured by atomic force microscopy, yielded expanded cells with higher proliferation capacity but lower chondrogenic potential when compared to those with varied durations of AA treatment. dECM with 250 µM of AA treatment for 10 d had better rejuvenation in chondrogenic capacity if the deposited cells were from passage 2 rather than passage 5, despite no significant difference in matrix stiffness. In the dose regimen study, we found that dECMs deposited by varied concentrations of AA yielded expanded cells with higher proliferation capacity despite lower expression levels of stem cell related surface markers. Compared to cells expanded on tissue culture polystyrene, those on dECM exhibited greater chondrogenic potential, particularly for the dECMs with 50 µM and 250 µM of AA treatment. With the supplementation of ethyl-3,4-dihydroxybenzoate (EDHB), an inhibitor targeting procollagen synthesis, the dECM with 50 µM of AA treatment exhibited a dramatic decrease in the rejuvenation effect of expanded cell chondrogenic potential at both mRNA and protein levels despite no significant difference in matrix stiffness. Defined AA treatments during matrix preparation will benefit dECM-mediated stem cell engineering and future treatments for cartilage defects.
Collapse
Affiliation(s)
- Tyler Pizzute
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Ying Zhang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
29
|
Farahzadi R, Mesbah-Namin SA, Zarghami N, Fathi E. L-carnitine Effectively Induces hTERT Gene Expression of Human Adipose Tissue-derived Mesenchymal Stem Cells Obtained from the Aged Subjects. Int J Stem Cells 2016; 9:107-14. [PMID: 27426092 PMCID: PMC4961110 DOI: 10.15283/ijsc.2016.9.1.107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2015] [Indexed: 11/22/2022] Open
Abstract
Background and Objectives Human mesenchymal stem cells (hMSCs) are attractive candidates for cell therapy and regenerative medicine due to their multipotency and ready availability, but their application can be complicated by the factors such as age of the donors and senescence-associated growth arrest during culture conditions. The latter most likely reflects the fact that aging of hMSCs is associated with a rise in intracellular reactive oxygen species, loss of telomerase activity, decrease in human telomerase reverse transcriptase (hTERT) expression and finally eroded telomere ends. Over-expression of telomerase in hMSCs leads to telomere elongation and may help to maintain replicative life–span of these cells. The aim of this study was to evaluate of the effect of L-carnitine (LC) as an antioxidant on the telomerase gene expression and telomere length in aged adipose tissue-derived hMSCs. Methods For this purpose, cells were isolated from healthy aged volunteers and their viabilities were assessed by MTT assay. Quantitative gene expression of hTERT and absolute telomere length measurement were also performed by real-time PCR in the absence and presence of different doses of LC (0.1, 0.2 and 0.4 mM). Results The results indicated that LC could significantly increase the hTERT gene expression and telomere length, especially in dose of 0.2 mM of LC and in 48 h treatment for the aged adipose tissue-derived hMSCs samples. Conclusion It seems that LC would be a good candidate to improve the lifespan of the aged adipose tissue-derived hMSCs due to over-expression of telomerase and lengthening of the telomeres.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tabriz, Iran
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
30
|
Nurkovic J, Dolicanin Z, Mustafic F, Mujanovic R, Memic M, Grbovic V, Skevin AJ, Nurkovic S. Mesenchymal stem cells in regenerative rehabilitation. J Phys Ther Sci 2016; 28:1943-8. [PMID: 27390452 PMCID: PMC4932093 DOI: 10.1589/jpts.28.1943] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/12/2016] [Indexed: 02/06/2023] Open
Abstract
[Purpose] Regenerative medicine and rehabilitation contribute in many ways to a specific
plan of care based on a patient’s medical status. The intrinsic self-renewing,
multipotent, regenerative, and immunosuppressive properties of mesenchymal stem cells
offer great promise in the treatment of numerous autoimmune, degenerative, and
graft-versus-host diseases, as well as tissue injuries. As such, mesenchymal stem cells
represent a therapeutic fortune in regenerative medicine. The aim of this review is to
discuss possibilities, limitations, and future clinical applications of mesenchymal stem
cells. [Subjects and Methods] The authors have identified and discussed clinically and
scientifically relevant articles from PubMed that have met the inclusion criteria.
[Results] Direct treatment of muscle injuries, stroke, damaged peripheral nerves, and
cartilage with mesenchymal stem cells has been demonstrated to be effective, with
synergies seen between cellular and physical therapies. Over the past few years, several
researchers, including us, have shown that there are certain limitations in the use of
mesenchymal stem cells. Aging and spontaneous malignant transformation of mesenchymal stem
cells significantly affect the functionality of these cells. [Conclusion] Definitive
conclusions cannot be made by these studies because limited numbers of patients were
included. Studies clarifying these results are expected in the near future.
Collapse
Affiliation(s)
- Jasmin Nurkovic
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia; Center for Physical Medicine and Rehabilitation, Clinical Center Kragujevac, Serbia; Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Zana Dolicanin
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia; General Hospital Novi Pazar, Serbia
| | | | - Rifat Mujanovic
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia
| | - Mensur Memic
- Department of Biomedical Sciences, State University of Novi Pazar, Serbia
| | - Vesna Grbovic
- Center for Physical Medicine and Rehabilitation, Clinical Center Kragujevac, Serbia; Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Aleksandra Jurisic Skevin
- Center for Physical Medicine and Rehabilitation, Clinical Center Kragujevac, Serbia; Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Selmina Nurkovic
- Faculty of Medical Sciences, University of Kragujevac, Serbia; General Hospital Novi Pazar, Serbia
| |
Collapse
|
31
|
Yao B, Huang S, Gao D, Xie J, Liu N, Fu X. Age-associated changes in regenerative capabilities of mesenchymal stem cell: impact on chronic wounds repair. Int Wound J 2015; 13:1252-1259. [PMID: 26424496 DOI: 10.1111/iwj.12491] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 06/30/2015] [Accepted: 08/07/2015] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent an ideal source of autologous cell-based therapy for chronic wounds. Functional characteristics of MSCs may benefit wound healing by exerting their multi-regenerative potential. However, cell ageing resulting from chronic degenerative diseases or donor age could cause inevitable effects on the regenerative abilities of MSCs. A variety of studies have shown the relationship between MSC ageing and age-related dysfunction, but few associate these age-related impacts on MSCs with their ability of repairing chronic wounds, which are common in the elderly population. Here, we discuss the age-associated changes of MSCs and describe the potential impacts on MSC-based therapy for chronic wounds. Furthermore, critical evaluation of the current literatures is necessary for understanding the underlying mechanisms of MSC ageing and raising the corresponding concerns on considering their possible use for chronic wound repair.
Collapse
Affiliation(s)
- Bin Yao
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China.,School of Medicine, Nankai University, Tianjin, China
| | - Sha Huang
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China.,Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing, P. R. China.,Hainan Branch of the Chinese PLA General Hospital, Sanya, P. R. China
| | - Dongyun Gao
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China.,Department of Oncology, Dongtai People's Hospital, Dongtai, P. R. China
| | - Jiangfan Xie
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China.,Graduate School of Tianjin Medical University, Tianjin, P. R. China
| | - Nanbo Liu
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China.,Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xiaobing Fu
- Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China.,Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing, P. R. China
| |
Collapse
|
32
|
Immortalisation with hTERT Impacts on Sulphated Glycosaminoglycan Secretion and Immunophenotype in a Variable and Cell Specific Manner. PLoS One 2015. [PMID: 26196672 PMCID: PMC4510558 DOI: 10.1371/journal.pone.0133745] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Limited options for the treatment of cartilage damage have driven the development of tissue engineered or cell therapy alternatives reliant on ex vivo cell expansion. The study of chondrogenesis in primary cells is difficult due to progressive cellular aging and senescence. Immortalisation via the reintroduction of the catalytic component of telomerase, hTERT, could allow repeated, longitudinal studies to be performed while bypassing senescent phenotypes. Methods Three human cell types: bone marrow-derived stromal cells (BMA13), embryonic stem cell-derived (1C6) and chondrocytes (OK3) were transduced with hTERT (BMA13H, 1C6H and OK3H) and proliferation, surface marker expression and tri-lineage differentiation capacity determined. The sulphated glycosaminoglycan (sGAG) content of the monolayer and spent media was quantified in maintenance media (MM) and pro-chondrogenic media (PChM) and normalised to DNA. Results hTERT expression was confirmed in transduced cells with proliferation enhancement in 1C6H and OK3H cells but not BMA13H. All cells were negative for leukocyte markers (CD19, CD34, CD45) and CD73 positive. CD14 was expressed at low levels on OK3 and OK3H and HLA-DR on BMA13 (84.8%). CD90 was high for BMA13 (84.9%) and OK3 (97.3%) and moderate for 1C6 (56.7%), expression was reduced in BMA13H (33.7%) and 1C6H (1.6%). CD105 levels varied (BMA13 87.7%, 1C6 8.2%, OK3 43.3%) and underwent reduction in OK3H (25.1%). 1C6 and BMA13 demonstrated osteogenic and adipogenic differentiation but mineralised matrix and lipid accumulation appeared reduced post hTERT transduction. Chondrogenic differentiation resulted in increased monolayer-associated sGAG in all primary cells and 1C6H (p<0.001), and BMA13H (p<0.05). In contrast OK3H demonstrated reduced monolayer-associated sGAG in PChM (p<0.001). Media-associated sGAG accounted for ≥55% (PChM-1C6) and ≥74% (MM-1C6H). Conclusion In conclusion, hTERT transduction could, but did not always, prevent senescence and cell phenotype, including differentiation potential, was affected in a variable manner. As such, these cells are not a direct substitute for primary cells in cartilage regeneration research.
Collapse
|
33
|
Chang TC, Hsu MF, Wu KK. High glucose induces bone marrow-derived mesenchymal stem cell senescence by upregulating autophagy. PLoS One 2015; 10:e0126537. [PMID: 25961745 PMCID: PMC4427318 DOI: 10.1371/journal.pone.0126537] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 04/02/2015] [Indexed: 02/07/2023] Open
Abstract
Hyperglycemia was reported to cause bone marrow hematopoietic niche dysfunction, and high glucose (HG) in the cultured medium induces MSC senescence. The underlying mechanism is unclear. Here, we investigated the role of HG-induced autophagy in bone-marrow-derived mesenchymal stem cell (BMSC) senescence. HG (25 mM) increased expression of Beclin-1, Atg 5, 7 and 12, generation of LC3-II and autophagosome formation which was correlated with development of cell senescence. Pretreatment of HG-MSC with 3-methyladenine (3-MA) prevented senescence but increased apoptosis. N-acetylcysteine (NAC) was effective in abrogating HG-induced autophagy accompanied by prevention of senescence. Diphenyleneiodonium (DPI), an inhibitor of NADPH oxidase, blocked autophagy and senescence in a manner comparable to NAC. 3-MA, NAC and DPI inhibited HG-induced interleukin-6 production in BMSCs. These results suggest that hyperglycemia induces MSC senescence and local inflammation via a novel oxidant-mediated autophagy which contributes to bone marrow niche dysfunction and hematopoietic impairment.
Collapse
Affiliation(s)
- Tzu-Ching Chang
- Metabolomic Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Min-Fen Hsu
- Metabolomic Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Kenneth K. Wu
- Metabolomic Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Basic Medicine, China Medical University, Taichung, Taiwan
- National Health Research Institutes, Zhunan, Taiwan
- * E-mail:
| |
Collapse
|
34
|
Human mesenchymal stem cells possess different biological characteristics but do not change their therapeutic potential when cultured in serum free medium. Stem Cell Res Ther 2014; 5:132. [PMID: 25476802 PMCID: PMC4445567 DOI: 10.1186/scrt522] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) are widely investigated in clinical researches to treat various diseases. Classic culture medium for MSCs, even for clinical use, contains fetal bovine serum. The serum-containing medium (SCM) seems a major obstacle for MSCs-related therapies due to the risk of contamination of infectious pathogens. Some studies showed that MSCs could be expanded in serum free medium (SFM); however, whether SFM would change the biological characteristics and safety issues of MSCs has not been well answered. Methods Human umbilical cord mesenchymal stem cells (hUC-MSCs) were cultured in a chemical defined serum free medium. Growth, multipotency, surface antigen expression, telomerase, immunosuppressive ability, gene expression profile and genomic stability of hUC-MSCs cultured in SFM and SCM were analyzed and compared side by side. Results hUC-MSCs propagated more slowly and senesce ultimately in SFM. SFM-expanded hUC-MSCs were different from SCM-expanded hUC-MSCs in growth rate, telomerase, gene expression profile. However, SFM-expanded hUC-MSCs maintained multipotency and the profile of surface antigen which were used to define human MSCs. Both SFM- and SCM-expanded hUC-MSCs gained copy number variation (CNV) in long-term in vitro culture. Conclusion hUC-MCSs could be expanded in SFM safely to obtain enough cells for clinical application, meeting the basic criteria for human mesenchymal stem cells. hUC-MSCs cultured in SFM were distinct from hUC-MSCs cultured in SCM, yet they remained therapeutic potentials for future regenerative medicine. Electronic supplementary material The online version of this article (doi:10.1186/scrt522) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Trend of telomerase activity change during human iPSC self-renewal and differentiation revealed by a quartz crystal microbalance based assay. Sci Rep 2014; 4:6978. [PMID: 25381797 PMCID: PMC4225532 DOI: 10.1038/srep06978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/20/2014] [Indexed: 11/09/2022] Open
Abstract
Telomerase plays an important role in governing the life span of cells for its capacity to extend telomeres. As high activity of telomerase has been found in stem cells and cancer cells specifically, various methods have been developed for the evaluation of telomerase activity. To overcome the time-consuming procedures and complicated manipulations of existing methods, we developed a novel method named Telomeric Repeat Elongation Assay based on Quartz crystal microbalance (TREAQ) to monitor telomerase activity during the self-renewal and differentiation of human induced pluripotent stem cells (hiPSCs). TREAQ results indicated hiPSCs possess invariable telomerase activity for 11 passages on Matrigel and a steady decline of telomerase activity when differentiated for different periods, which is confirmed with existing golden standard method. The pluripotency of hiPSCs during differentiation could be estimated through monitoring telomerase activity and compared with the expression levels of markers of pluripotency gene via quantitative real time PCR. Regular assessment for factors associated with pluripotency or stemness was expensive and requires excessive sample consuming, thus TREAQ could be a promising alternative technology for routine monitoring of telomerase activity and estimate the pluripotency of stem cells.
Collapse
|
36
|
Wilson B, Novakofski KD, Donocoff RS, Liang YXA, Fortier LA. Telomerase Activity in Articular Chondrocytes Is Lost after Puberty. Cartilage 2014; 5:215-20. [PMID: 26069700 PMCID: PMC4335769 DOI: 10.1177/1947603514537518] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Telomere length and telomerase activity are important indicators of cellular senescence and replicative ability. Loss of telomerase is associated with ageing and the development of osteoarthritis. Implantation of telomerase-positive cells, chondrocytes, or stem cells expressing a normal chondrocyte phenotype is desired for cartilage repair procedures. The objective of this study was to identify at what age chondrocytes and at what passage bone marrow-derived mesenchymal stem cells (MSCs) become senescent based on telomerase activity. The effect of osteogenic protein-1 (OP-1) or interleukin-1α (IL-1α) treatment on telomerase activity in chondrocytes was also measured to determine the response to anabolic or catabolic stimuli. METHODS Articular cartilage was collected from horses (n = 12) aged 1 month to 18 years. Chondrocytes from prepubescent horses (<15 months) were treated with OP-1 or IL-1α. Bone marrow aspirate from adult horses was collected and cultured for up to 10 days to isolate MSCs. Telomerase activity was measured using the TeloTAGGG Telomerase PCR ELISA kit. RESULTS Chondrocytes from prepubescent horses were positive for telomerase activity. Treatment with IL-1α resulted in a decrease in chondrocyte telomerase activity; however, treatment with OP-1 did not change telomerase activity. One MSC culture sample was positive for telomerase activity on day 2; all samples were negative for telomerase activity on day 10. CONCLUSIONS These results suggest that chondrocytes from prepubescent donors are potentially more suitable for cartilage repair procedures and that telomerase activity is diminished by anabolic and catabolic cytokine stimulation. If MSCs are utilized in cartilage repair, minimal passaging should be performed prior to implantation.
Collapse
Affiliation(s)
- Brooke Wilson
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA
| | | | | | | | - Lisa A. Fortier
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|
37
|
Bellayr IH, Catalano JG, Lababidi S, Yang AX, Lo Surdo JL, Bauer SR, Puri RK. Gene markers of cellular aging in human multipotent stromal cells in culture. Stem Cell Res Ther 2014; 5:59. [PMID: 24780490 PMCID: PMC4055144 DOI: 10.1186/scrt448] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/15/2014] [Indexed: 12/14/2022] Open
Abstract
Introduction Human multipotent stromal cells (MSCs) isolated from bone marrow or other tissue sources have great potential to treat a wide range of injuries and disorders in the field of regenerative medicine and tissue engineering. In particular, MSCs have inherent characteristics to suppress the immune system and are being studied in clinical studies to prevent graft-versus-host disease. MSCs can be expanded in vitro and have potential for differentiation into multiple cell lineages. However, the impact of cell passaging on gene expression and function of the cells has not been determined. Methods Commercially available human MSCs derived from bone marrow from six different donors, grown under identical culture conditions and harvested at cell passages 3, 5, and 7, were analyzed with gene-expression profiling by using microarray technology. Results The phenotype of these cells did not change as reported previously; however, a statistical analysis revealed a set of 78 significant genes that were distinguishable in expression between passages 3 and 7. None of these significant genes corresponded to the markers established by the International Society for Cellular Therapy (ISCT) for MSC identification. When the significant gene lists were analyzed through pathway analysis, these genes were involved in the top-scoring networks of cellular growth and proliferation and cellular development. A meta-analysis of the literature for significant genes revealed that the MSCs seem to be undergoing differentiation into a senescent cell type when cultured extensively. Consistent with the differences in gene expression at passage 3 and 7, MSCs exhibited a significantly greater potential for cell division at passage 3 in comparison to passage 7. Conclusions Our results identified specific gene markers that distinguish aging MSCs grown in cell culture. Confirmatory studies are needed to correlate these molecular markers with biologic attributes that may facilitate the development of assays to test the quality of MSCs before clinical use.
Collapse
|
38
|
Saeed H, Iqtedar M. Stem cell function and maintenance - ends that matter: role of telomeres and telomerase. J Biosci 2014; 38:641-9. [PMID: 23938394 DOI: 10.1007/s12038-013-9346-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stem cell research holds a promise to treat and prevent age-related degenerative changes in humans. Literature is replete with studies showing that stem cell function declines with aging, especially in highly proliferative tissues/ organs. Among others, telomerase and telomere damage is one of the intrinsic physical instigators that drive agerelated degenerative changes. In this review we provide brief overview of telomerase-deficient aging affects in diverse stem cells populations. Furthermore, potential disease phenotypes associated with telomerase dysregulation in a specific stem cell population is also discussed in this review. Additionally, the role of telomerase in stem cell driven cancer is also briefly touched upon.
Collapse
Affiliation(s)
- Hamid Saeed
- Department of Endocrinology, School of Medicine, Stanford University, Stanford, CA, USA.
| | | |
Collapse
|
39
|
Telomere length analysis of human mesenchymal stem cells by quantitative PCR. Gene 2013; 519:348-55. [PMID: 23380569 DOI: 10.1016/j.gene.2013.01.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/16/2013] [Indexed: 12/11/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have attracted much attention for tissue repair and wound healing because of their self-renewal capacity and multipotentiality. In order to mediate an effective therapy, substantial numbers of cells are required, which necessitates extensive sub-culturing and expansion of hMSCs. Throughout ex vivo expansion, the cells undergo telomere shortening, and critically short telomeres can trigger loss of cell viability. Telomeres are nucleoprotein structures that cap the ends of chromosomes, and serve to protect the DNA from the degradation which occurs due to the end-replication problem in all eukaryotes. As hMSCs have only a finite ability for self-renewal like most somatic cells, assaying for telomere length in hMSCs provides critical information on the replicative capacity of the cells, an important criterion in the selection of hMSCs for therapy. Telomere length is generally quantified by Southern blotting and fluorescence in situ hybridization, and more recently by PCR-based methods. Here we describe the quantification of hMSC telomere length by real-time PCR; our results demonstrate the effect of telomere shortening on the proliferation and clonogenicity of hMSCs. Thus, this assay constitutes a useful tool for the determination of relative telomere length in hMSCs.
Collapse
|
40
|
Piper SL, Wang M, Yamamoto A, Malek F, Luu A, Kuo AC, Kim HT. Inducible immortality in hTERT-human mesenchymal stem cells. J Orthop Res 2012; 30:1879-85. [PMID: 22674533 DOI: 10.1002/jor.22162] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 05/09/2012] [Indexed: 02/04/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are attractive candidates for tissue engineering and cell-based therapy because of their multipotentiality and availability in adult donors. However, in vitro expansion and differentiation of these cells is limited by replicative senescence. The proliferative capacity of hMSCs can be enhanced by ectopic expression of telomerase, allowing for long-term culture. However, hMSCs with constitutive telomerase expression demonstrate unregulated growth and even tumor formation. To address this problem, we used an inducible Tet-On gene expression system to create hMSCs in which ectopic telomerase expression can be induced selectively by the addition of doxycycline (i-hTERT hMSCs). i-hTERT hMSCs have inducible hTERT expression and telomerase activity, and are able to proliferate significantly longer than wild type hMSCs when hTERT expression is induced. They stop proliferating when hTERT expression is turned off and can be rescued when expression is re-induced. They retain multipotentiality in vitro even at an advanced age. We also used a selective inhibitor of telomere elongation to show that the mechanism driving immortalization of hMSCs by hTERT is dependent upon maintenance of telomere length. Thanks to their extended lifespan, preserved multipotentiality and controlled growth, i-hTERT hMSCs may prove to be a useful tool for the development and testing of novel stem cell therapies.
Collapse
Affiliation(s)
- Samantha L Piper
- Department of Orthopaedic Surgery, University of California San Francisco, 500 Parnassus Avenue, San Francisco, California 94143, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
DNA damage, discoordinated gene expression and cellular senescence in osteoarthritic chondrocytes. Osteoarthritis Cartilage 2012; 20:1020-8. [PMID: 22659602 DOI: 10.1016/j.joca.2012.05.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 05/15/2012] [Accepted: 05/23/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The initiation/progression factors of osteoarthritic (OA) cartilage degeneration and the involved biological mechanisms remain rather enigmatic. One core reason for this might be a cellular senescence-like phenotype of OA chondrocytes, which might show a fundamentally different behavior pattern unexpected from the biological mechanism established in young cells. DESIGN This study was designed to investigate one core property of senescent cells, the heterogeneity of gene expression, in OA chondrocytes by double-labeling immunolocalization using two genes (vimentin, S-100 protein) as surrogates, which are constitutively expressed by (normal) chondrocytes. The level of genomic DNA damage in OA chondrocytes was compared to normal chondrocytes and in vitro experiments designed to demonstrate that stochastic genomic DNA damage is able to induce heterogeneity of gene expression in chondrocytes. RESULTS We show a significantly increased heterogeneity of gene expression for vimentin and S-100 protein as well as a significantly increased genomic DNA damage in the OA compared to normal chondrocytes, whereas no evidence of critical telomere shortening was found. In vitro experiments demonstrated that stochastic genomic DNA damage induced by increased oxidative or genotoxic stress is able to induce the heterogeneity in gene expression found in the OA cells in situ. CONCLUSIONS Our results suggest that OA chondrocytes show a special form of age-related cell degeneration, "progressive/stress-induced senescence", progressing over time due to accumulated DNA damage and subsequent chaotic gene activation pattern. This promotes increased malfunctioning of the cells and finally the loss of their capacity to keep up cell and tissue homeostasis, i.e., prevent OA.
Collapse
|
42
|
Li J, Pei M. Cell Senescence: A Challenge in Cartilage Engineering and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:270-87. [PMID: 22273114 DOI: 10.1089/ten.teb.2011.0583] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
- Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
- Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
43
|
Righolt CH, van 't Hoff MLR, Vermolen BJ, Young IT, Raz V. Robust nuclear lamina-based cell classification of aging and senescent cells. Aging (Albany NY) 2012; 3:1192-201. [PMID: 22199022 PMCID: PMC3273899 DOI: 10.18632/aging.100414] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Changes in the shape of the nuclear lamina are exhibited in senescent cells, as well as in cells expressing mutations in lamina genes. To identify cells with defects in the nuclear lamina we developed an imaging method that quantifies the intensity and curvature of the nuclear lamina. We show that this method accurately describes changes in the nuclear lamina. Spatial changes in nuclear lamina coincide with redistribution of lamin A proteins and local reduction in protein mobility in senescent cell. We suggest that local accumulation of lamin A in the nuclear envelope leads to bending of the structure. A quantitative distinction of the nuclear lamina shape in cell populations was found between fresh and senescent cells, and between primary myoblasts from young and old donors. Moreover, with this method mutations in lamina genes were significantly distinct from cells with wild-type genes. We suggest that this method can be applied to identify abnormal cells during aging, in in vitro propagation, and in lamina disorders.
Collapse
|
44
|
Kim M, Kim C, Choi YS, Kim M, Park C, Suh Y. Age-related alterations in mesenchymal stem cells related to shift in differentiation from osteogenic to adipogenic potential: implication to age-associated bone diseases and defects. Mech Ageing Dev 2012; 133:215-25. [PMID: 22738657 DOI: 10.1016/j.mad.2012.03.014] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Revised: 03/17/2012] [Accepted: 03/28/2012] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cells (MSC) have attracted considerable attention in the fields of cell and gene therapy due to their intrinsic ability to differentiate into multiple lineages. The various therapeutic applications involving MSC require initial expansion and/or differentiation in vitro prior to clinical use. However, serial passages of MSC in culture lead to decreased differentiation potential and stem cell characteristics, eventually inducing cellular aging which will limit the success of cell-based therapeutic interventions. Here we review the age-related changes that occur in MSC with a special focus on the shift of differentiation potential from osteogenic to adipogenic lineage during the MSC aging processes and how aging causes this preferential shift by oxidative stress and/or energy metabolism defect. Oxidative stress-related signals and some microRNAs affect the differentiation potential shift of MSC by directly targeting key regulatory factors such as Runx-2 or PPAR-γ, and energy metabolism pathway is involved as well. All information described here including transcription factors, microRNAs and FoxOs could be used towards development of treatment regimens for age-related bone diseases and related defects based on mutually exclusive lineage fate determination of MSC.
Collapse
Affiliation(s)
- MiJung Kim
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Jeon BG, Kwack DO, Rho GJ. Variation of telomerase activity and morphology in porcine mesenchymal stem cells and fibroblasts during prolonged in vitro culture. Anim Biotechnol 2012; 22:197-210. [PMID: 22132813 DOI: 10.1080/10495398.2011.624651] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The purpose of this study was to examine the telomerase activity, population doubling time (PDT), morphological alterations, and the cell cycle status with activity of senescence-associated-ß-galactosidase in porcine mesenchymal stem cells (MSCs) and fibroblasts during an extended in vitro culture. MSCs and fibroblasts were isolated from bone marrow and ear skin of a miniature pig, respectively, and cultured up to 20 passages. The analysis was carried out in MSCs and fibroblasts at 1, 5, 10, 15, and 20 passages. Relative telomerase activity (RTA) levels were significantly (P < 0.05) higher in MSCs than in fibroblasts at all the passages. The PDT and cellular size slightly increased in MSCs at later passages. In contrast, fibroblasts had significantly (P < 0.05) increased PDT and cellular size, and the morphology revealed senescent-like abnormal type after passage 10. Further, the high incidence of ß-galactosidase stained cells was observed in fibroblasts compared to that of MSCs at passage 15, and cell cycle stage at G0 / G1 phase was significantly (P < 0.05) increased in the fibroblasts at 15 and 20 passages compared to that of MSCs. Based on these observations, we concluded that porcine MSCs possessed more tolerance against senescence and aging compared to fibroblasts following prolonged in vitro culture.
Collapse
Affiliation(s)
- Byeong-Gyun Jeon
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | | | | |
Collapse
|
46
|
Neri S, Mariani E, Cattini L, Facchini A. Long-term in vitro expansion of osteoarthritic human articular chondrocytes do not alter genetic stability: a microsatellite instability analysis. J Cell Physiol 2011; 226:2579-85. [PMID: 21792915 DOI: 10.1002/jcp.22603] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this study, we investigated genetic damage acquisition during in vitro culture of human osteoarthritic (OA) chondrocytes to evaluate their safety for use in regenerative medicine clinical applications. In particular, we have addressed the impact of long-term in vitro culture on simple sequence repeat stability, to evaluate the involvement of the mismatch repair system (MMR) in the accumulation of genetic damage. MMR, the main post-replicative correction pathway, has a fundamental role in maintaining genomic stability and can be monitored by assessing microsatellite instability (MSI). MMR activity has been reported to decrease with age not only in vivo, but also in vitro in relationship to culture passages. OA chondrocytes from seven donors were cultured corresponding to 13-29 population doublings. Aliquots of the cells were collected and analyzed for MSI at five DNA loci (CD4, VWA, FES, TPOX, and P53) and for MMR gene expression at each subculture. Genetic stability was confirmed throughout the culture period. MMR genes demonstrated a strong coordination at the transcriptional level among the different components; expression levels were very low, in accordance with the observed genetic stability. The reduced expression of MMR genes might underline no need for increasing DNA repair control in the culture conditions tested, in which no genetic damage was evidenced. These data argue for the safety of chondrocytes for cellular therapies and are encouraging for the potential use of in vitro expanded OA chondrocytes, supporting the extension of autologous cell therapy procedures to degenerative articular diseases.
Collapse
Affiliation(s)
- Simona Neri
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto di Ricerca Codivilla Putti, Istituto Ortopedico Rizzoli, Bologna, Italy.
| | | | | | | |
Collapse
|
47
|
Breu A, Sprinzing B, Merkl K, Bechmann V, Kujat R, Jenei-Lanzl Z, Prantl L, Angele P. Estrogen reduces cellular aging in human mesenchymal stem cells and chondrocytes. J Orthop Res 2011; 29:1563-71. [PMID: 21469181 DOI: 10.1002/jor.21424] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Accepted: 03/10/2011] [Indexed: 02/04/2023]
Abstract
Chondrocyte aging is associated with cartilage degeneration and senescence impairs the regenerative potential of mesenchymal stem cells (MSCs). Estrogen exerts profound effects on human physiology including articular cartilage and MSCs. The present study should analyze the effects of pre- and postmenopausal estrogen concentrations on chondrogenic cells. Physiologic premenopausal concentrations of 17β-estradiol (E(2)) significantly decelerated telomere attrition in MSCs and chondrocytes while postmenopausal E(2) concentration had no significant effects. The estrogen agonist-antagonist tamoxifen did not affect telomere biology, but inhibited the E(2) -stimulated reduction in telomere shortening. E(2) and tamoxifen did not influence cell proliferation, cell morphology, and β-galactosidase staining in chondrogenic cells. E(2) treatment did not affect the telomere-associated proteins TRF1 and TRF2. E(2) had no regulatory effects on the expression rates of the cell cycle regulator p21 and the DNA repair proteins SIRT1 and XRCC5. In spite of reducing telomere shortening in aging MSCs and chondrocytes, estrogen is not able to prevent somatic cells from replicative exhaustion and from finally entering senescence. The fade of telomere shortening under pre- to postmenopausal estrogen concentrations suggests, at least in part, a senescence-dependent cause for the onset of osteoarthritis in women after menopause.
Collapse
Affiliation(s)
- Anita Breu
- Department of Anesthesiology, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93042 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Tan Q, Lui PPY, Rui YF. Effect of in vitro passaging on the stem cell-related properties of tendon-derived stem cells-implications in tissue engineering. Stem Cells Dev 2011; 21:790-800. [PMID: 21627568 DOI: 10.1089/scd.2011.0160] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This study aimed to compare clonogenicity, proliferation, stem cell-related marker expression, senescence, and differentiation potential of rat patellar tendon-derived stem cells (TDSCs) at early (P5), mid (P10), and late (P20, P30) passages. The clonogenicity of the cells was assessed by colony-forming assay and their proliferative potential was assessed by bromodeoxyuridine assay. The surface expression of CD90 and CD73 was assessed by flow cytometry. The cellular senescence was assessed by β-galactosidase activity. The adipogenic, chondrogenic, and osteogenic differentiation potentials of TDSCs were assessed by standard assays after induction. The mRNA expression of tendon-related markers, scleraxis (Scx) and tenomodulin (Tnmd), was measured by quantitative real-time reverse transcription-polymerase chain reaction. Both the colony numbers and proliferative potential of TDSCs increased with passaging. Concomitantly, there was significant upregulation of β-galactosidase activity with TDSC passaging. The subculture of TDSCs downregulated the expression of CD90 and CD73. Lipid droplets were formed in the early and mid passages of TDSCs upon adipogenic induction, but were absent in the late passages. The expression of peroxisome proliferator activator receptor gamma 2 (PPARγ2) and CCAAT/enhancer binding protein alpha (C/EBPα) in TDSCs after adipogenic induction decreased with passaging. Chondrogenesis, proteoglycan deposition, collagen type II protein expression, collagen type 2A1 (Col2AI), and aggrecan (Acan) mRNA expression were less in pellets formed with later passages of TDSCs after chondrogenic induction. The expression of Scx and Tnmd was lower in the late, compared with early and mid, passages of TDSCs. However, matrix mineralization and expression of alkaline phosphatase (Alpl) and osteocalcin (Bglap) mRNA after osteogenic induction increased with TDSC passaging. Researchers and clinicians should consider the changes of stem cell-related properties of TDSCs when multiplying them in vitro for tissue engineering.
Collapse
Affiliation(s)
- Qi Tan
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | | |
Collapse
|
49
|
Characterization and comparison of telomere length, telomerase and reverse transcriptase activity and gene expression in human mesenchymal stem cells and cancer cells of various origins. Cell Tissue Res 2011; 345:149-61. [PMID: 21638208 DOI: 10.1007/s00441-011-1191-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 05/12/2011] [Indexed: 02/06/2023]
Abstract
We have characterized and compared the telomere length, telomerase, reverse transcriptase (RT) activity and expression of genes implicated in cancer and in pluripotency, in human mesenchymal stem cells (MSCs) derived from dental papilla tissue, umbilical cord matrix and adipose tissue and in cancer cells (MDA-MB-231, U-87 MG, and MCF-7). MRC-5 fetal fibroblasts and adult muscle cells were used as somatic cell controls. Telomere length was significantly (P<0.05) higher in MSCs and somatic cells (7.2-9.3 kb) than in cancer cell lines (3.9-6 kb). However, the relative telomerase activity (RTA) in the cancer cell lines was significantly (P<0.05) higher than that of MSCs and somatic cells. RTA tended to be slightly higher in MSCs but no significant differences were observed between some cancer cells and MSCs. However, RTA was not detected in somatic cells. Although differentially displayed, the expression of genes related to cancer (BCL-2, p53, NF-κB, TGF-β, VEGF) and transcription and pluripotency (OCT4, NANOG, STAT3, REX1) were commonly observed in MSCs and cancer cells. Thus, endogenous non-telomerase RTA might be a potential biological marker or regulator among MSCs and cancer cells. Further, by sharing the biological and molecular markers of self-renewal and proliferation with cancer cells, MSCs might play a contributory role as tissue resident stem cells in tumor development.
Collapse
|
50
|
Griffiths SL, Griffiths D, Forsyth NR. Mesenchymal stem-cell-based therapy for the older patient. ACTA ACUST UNITED AC 2011. [DOI: 10.2217/ahe.11.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bone-marrow aspirate-derived mesenchymal stem cells (MSCs) form a core element of the vast majority of adherent, autologous stem-cell-based therapeutic clinical trials. This article explores the existing literature to determine if proposed MSC-based autologous cell therapy is suitable for use in an elderly patient population. Within the review a number of questions are asked: does the bone-marrow aspirate mononuclear cell (MNC) number, the frequency of MSC colony forming units or their proliferative potential correlate with age? Does disease state have an additional impact? Are immunophenotypes, molecular profiles or differentiation capacity linked to donor age? The answers to the above questions are essential for determining the appropriateness of elderly patients as donors and recipients for autologous MSC-based trials. Taken together, MSC yield, MNC number and proliferative potential all decrease with age while differentiation potential appears unchanged. The retention of differentiation capacity suggests that the elderly patient may be suitable for MSC-based therapy; however, only when isolation protocols are tailored in an individual-specific manner to correct for decreases in MNC number and MSC yield.
Collapse
Affiliation(s)
- Sarah L Griffiths
- The Guy Hilton Research Laboratories, Institute of Science & Technology in Medicine, Keele University Medical School, Stoke-on-Trent, ST4 7QB, UK
| | - David Griffiths
- Department of Orthopaedics, City General Hospital, Stoke-on-Trent, ST4 6QG, UK
| | | |
Collapse
|