1
|
Di Pietro E, Burla R, La Torre M, González-García MP, Dello Ioio R, Saggio I. Telomeres: an organized string linking plants and mammals. Biol Direct 2024; 19:119. [PMID: 39568075 PMCID: PMC11577926 DOI: 10.1186/s13062-024-00558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024] Open
Abstract
Telomeres are pivotal determinants of cell stemness, organismal aging, and lifespan. Herein, we examined similarities in telomeres of Arabidopsis thaliana, mice, and humans. We report the common traits, which include their composition in multimers of TTAGGG sequences and their protection by specialized proteins. Moreover, given the link between telomeres, on the one hand, and cell proliferation and stemness on the other, we discuss the counterintuitive convergence between plants and mammals in this regard, focusing on the impact of niches on cell stemness. Finally, we suggest that tackling the study of telomere function and cell stemness by taking into consideration both plants and mammals can aid in the understanding of interconnections and contribute to research focusing on aging and organismal lifespan determinants.
Collapse
Affiliation(s)
- Edison Di Pietro
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy
| | - Romina Burla
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy
- CNR Institute of Biology and Pathology, Rome, Italy
| | - Mattia La Torre
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy
| | - Mary-Paz González-García
- Centro de Biotecnología y Genómica de Plantas (Universidad Politécnica de Madrid - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria), UPM-INIA/CSIC. Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Raffaele Dello Ioio
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy.
| | - Isabella Saggio
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza, University of Rome, Rome, Italy.
| |
Collapse
|
2
|
Arellano MYG, VanHeest M, Emmadi S, Abdul-Hafez A, Ibrahim SA, Thiruvenkataramani RP, Teleb RS, Omar H, Kesaraju T, Mohamed T, Madhukar BV, Omar SA. Role of Mesenchymal Stem/Stromal Cells (MSCs) and MSC-Derived Extracellular Vesicles (EVs) in Prevention of Telomere Length Shortening, Cellular Senescence, and Accelerated Biological Aging. Bioengineering (Basel) 2024; 11:524. [PMID: 38927760 PMCID: PMC11200821 DOI: 10.3390/bioengineering11060524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Biological aging is defined as a progressive decline in tissue function that eventually results in cell death. Accelerated biologic aging results when the telomere length is shortened prematurely secondary to damage from biological or environmental stressors, leading to a defective reparative mechanism. Stem cells therapy may have a potential role in influencing (counteract/ameliorate) biological aging and maintaining the function of the organism. Mesenchymal stem cells, also called mesenchymal stromal cells (MSCs) are multipotent stem cells of mesodermal origin that can differentiate into other types of cells, such as adipocytes, chondrocytes, and osteocytes. MSCs influence resident cells through the secretion of paracrine bioactive components such as cytokines and extracellular vesicles (EVs). This review examines the changes in telomere length, cellular senescence, and normal biological age, as well as the factors contributing to telomere shortening and accelerated biological aging. The role of MSCs-especially those derived from gestational tissues-in prevention of telomere shortening (TS) and accelerated biological aging is explored. In addition, the strategies to prevent MSC senescence and improve the antiaging therapeutic application of MSCs and MSC-derived EVs in influencing telomere length and cellular senescence are reviewed.
Collapse
Affiliation(s)
- Myrna Y. Gonzalez Arellano
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Matthew VanHeest
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sravya Emmadi
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Amal Abdul-Hafez
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sherif Abdelfattah Ibrahim
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ranga P. Thiruvenkataramani
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Rasha S. Teleb
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Department of Pediatrics and Neonatology, Qena Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hady Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tulasi Kesaraju
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tarek Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Burra V. Madhukar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Said A. Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| |
Collapse
|
3
|
Mavinga M, Palmier M, Rémy M, Jeannière C, Lenoir S, Rey S, Saint-Marc M, Alonso F, Génot E, Thébaud N, Chevret E, Mournetas V, Rousseau B, Boiziau C, Boeuf H. The Journey of SCAPs (Stem Cells from Apical Papilla), from Their Native Tissue to Grafting: Impact of Oxygen Concentration. Cells 2022; 11:cells11244098. [PMID: 36552862 PMCID: PMC9776846 DOI: 10.3390/cells11244098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Tissue engineering strategies aim at characterizing and at optimizing the cellular component that is combined with biomaterials, for improved tissue regeneration. Here, we present the immunoMap of apical papilla, the native tissue from which SCAPs are derived. We characterized stem cell niches that correspond to a minority population of cells expressing Mesenchymal stromal/Stem Cell (CD90, CD105, CD146) and stemness (SSEA4 and CD49f) markers as well as endothelial cell markers (VWF, CD31). Based on the colocalization of TKS5 and cortactin markers, we detected migration-associated organelles, podosomes-like structures, in specific regions and, for the first time, in association with stem cell niches in normal tissue. From six healthy teenager volunteers, each with two teeth, we derived twelve cell banks, isolated and amplified under 21 or 3% O2. We confirmed a proliferative advantage of all banks when cultured under 3% versus 21% O2. Interestingly, telomerase activity was similar to that of the highly proliferative hiPSC cell line, but unrelated to O2 concentration. Finally, SCAPs embedded in a thixotropic hydrogel and implanted subcutaneously in immunodeficient mice were protected from cell death with a slightly greater advantage for cells preconditioned at 3% O2.
Collapse
Affiliation(s)
- Marine Mavinga
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | | | - Murielle Rémy
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | | | - Solène Lenoir
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Sylvie Rey
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | | | - Florian Alonso
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Elisabeth Génot
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Noélie Thébaud
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
| | - Edith Chevret
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France
| | | | - Benoit Rousseau
- Univ. Bordeaux, Animal Facility A2, Service Commun des Animaleries, F-33000 Bordeaux, France
| | | | - Helene Boeuf
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
4
|
Effect of Human Platelet Lysate as Cultivation Nutrient Supplement on Human Natal Dental Pulp Stem Cell In Vitro Expansion. Biomolecules 2022; 12:biom12081091. [PMID: 36008985 PMCID: PMC9405745 DOI: 10.3390/biom12081091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/29/2022] Open
Abstract
Despite several scientific or ethical issues, fetal bovine serum (FBS) remains the standard nutrient supplement in the mesenchymal stem cell cultivation medium. Cell amplification plays an important role in human stem cell therapies. Increasing interest in this field has supported attempts to find suitable human alternatives to FBS for in vitro cell propagation. Human platelet lysate (hPL) has recently been determined as one of them. Our study aimed to evaluate the influence of 2% hPL in the growth medium for in vitro expansion of human natal dental pulp stem cells (hNDP-SCs). The effect was determined on proliferation rate, viability, phenotype profile, expression of several markers, relative telomere length change, and differentiation potential of four lineages of hNDP-SCs. As a control, hNDP-SCs were simultaneously cultivated in 2% FBS. hNDP-SCs cultivated in hPL showed a statistically significantly higher proliferation rate in initial passages. We did not observe a statistically significant effect on mesenchymal stem cell marker (CD29, CD44, CD73, CD90) or stromal-associated marker (CD13, CD166) expression. The cell viability, relative telomere length, or multipotency remained unaffected in hNDP-SCs cultivated in hPL-medium. In conclusion, hPL produced under controlled and standardized conditions is an efficient serum supplement for in vitro expansion of hNDP-SCs.
Collapse
|
5
|
Dental Pulp Stem Cell Heterogeneity: Finding Superior Quality "Needles" in a Dental Pulpal "Haystack" for Regenerative Medicine-Based Applications. Stem Cells Int 2022; 2022:9127074. [PMID: 35027930 PMCID: PMC8752304 DOI: 10.1155/2022/9127074] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
Human dental pulp stem/stromal cells (hDPSCs) derived from the permanent secondary dentition are recognised to possess certain advantageous traits, which support their potential use as a viable source of mesenchymal stem/stromal cells (MSCs) for regenerative medicine-based applications. However, the well-established heterogeneous nature of hDPSC subpopulations, coupled with their limited numbers within dental pulp tissues, has impeded our understanding of hDPSC biology and the translation of sufficient quantities of these cells from laboratory research, through successful therapy development and clinical applications. This article reviews our current understanding of hDPSC biology and the evidence underpinning the molecular basis of their heterogeneity, which may be exploited to distinguish individual subpopulations with specific or superior characteristics for regenerative medicine applications. Pertinent unanswered questions which still remain, regarding the developmental origins, hierarchical organisation, and stem cell niche locations of hDPSC subpopulations and their roles in hDPSC heterogeneity and functions, will further be explored. Ultimately, a greater understanding of how key features, such as specific cell surface, senescence and other relevant genes, and protein and metabolic markers, delineate between hDPSC subpopulations with contrasting stemness, proliferative, multipotency, immunomodulatory, anti-inflammatory, and other relevant properties is required. Such knowledge advancements will undoubtedly lead to the development of novel screening, isolation, and purification strategies, permitting the routine and effective identification, enrichment, and expansion of more desirable hDPSC subpopulations for regenerative medicine-based applications. Furthermore, such innovative measures could lead to improved cell expansion, manufacture, and banking procedures, thereby supporting the translational development of hDPSC-based therapies in the future.
Collapse
|
6
|
Shetty A, Krishna M, Manjappa A, Shetty V, Hegde M, Kumar B. Comparative characterization and analysis of telomere length in stem cells derived from deciduous and permanent teeth. Dent Res J (Isfahan) 2022. [DOI: 10.4103/1735-3327.353833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
7
|
Strategies to Improve the Efficiency of Transplantation with Mesenchymal Stem Cells for the Treatment of Ischemic Stroke: A Review of Recent Progress. Stem Cells Int 2021; 2021:9929128. [PMID: 34490053 PMCID: PMC8418553 DOI: 10.1155/2021/9929128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022] Open
Abstract
Cerebral ischemia is a common global disease that is characterized by a loss of neurological function and a poor prognosis in many patients. However, only a limited number of treatments are available for this condition at present. Given that the efficacies of these treatments tend to be poor, cerebral ischemia can create a significant burden on patients, families, and society. Mesenchymal stem cell (MSC) transplantation treatment has shown significant potential in animal models of ischemic stroke; however, the specific mechanisms underlying this effect have yet to be elucidated. Furthermore, clinical trials have yet to yield promising results. Consequently, there is an urgent need to identify new methods to improve the efficiency of MSC transplantation as an optimal treatment for ischemic stroke. In this review, we provide an overview of recent scientific reports concerning novel strategies that promote MSC transplantation as an effective therapeutic approach, including physical approaches, chemical agents, traditional Chinese medicines and extracts, and genetic modification. Our analyses showed that two key factors need to be considered if we are to improve the efficacy of MSC transplantation treatments: survival ability and homing ability. We also highlight the importance of other significant mechanisms, including the enhanced activation of MSCs to promote neurogenesis and angiogenesis, and the regulation of permeability in the blood-brain barrier. Further in-depth investigations of the specific mechanisms underlying MSC transplantation treatment will help us to identify effective methods that improve the efficiency of MSC transplantation for ischemic stroke. The development of safer and more effective methods will facilitate the application of MSC transplantation as a promising adjuvant therapy for the treatment of poststroke brain damage.
Collapse
|
8
|
Zhang T, Ding H, Wang Y, Yuan Z, Zhang Y, Chen G, Xu Y, Chen L. Akt3-mTOR regulates hippocampal neurogenesis in adult mouse. J Neurochem 2021; 159:498-511. [PMID: 34077553 DOI: 10.1111/jnc.15441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/16/2021] [Accepted: 05/26/2021] [Indexed: 01/19/2023]
Abstract
Akt signaling has been associated with adult neurogenesis in the hippocampal dentate gyrus (DG). We reported cognitive dysfunction in Akt3 knockout (Akt3-KO) mice with the down-regulation of mTOR activation. However, little is known about the effects of Akt3 signaling on hippocampal neurogenesis. Herein, we show that progenitor cells, neuroblasts, and mature newborn neurons in hippocampal DG expressed Akt3 protein. The Akt3 phosphorylation in hippocampal DG was increased after voluntary wheel running for 7 days in wild-type mice (running WT mice), but not in Akt3-KO mice (running Akt3-KO mice). Subsequently, we observed that the proliferation of progenitor cells was suppressed in Akt3-KO mice and the mTOR inhibitor rapamycin-treated mice, whereas enhanced in running WT mice rather than running Akt3-KO mice. Neurite growth of neuroblasts was impaired in Akt3-KO mice and rapamycin-treated mice. In contrast, neither differentiation of progenitor cells nor migrating of newly generated neurons was altered in Akt3-KO mice or running WT mice. The levels of p70S6K and 4EBP1 phosphorylation were declined in Akt3-KO mice and elevated in running WT mice depending on mTOR activation. Furthermore, telomerase activity, telomere length, and expression of telomerase reverse transcriptase (TERT) were decreased in Akt3-KO mice but increased in running WT mice rather than running Akt3-KO mice, which required the mTOR activation. The study provides in vivo evidence that Akt3-mTOR signaling plays an important role in the proliferation of progenitor cells and neurite growth through positive regulated TERT expression and activation of p70S6K and 4EBP1.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Hong Ding
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Zihao Yuan
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yajie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
The Effect of Cultivation Passaging on the Relative Telomere Length and Proliferation Capacity of Dental Pulp Stem Cells. Biomolecules 2021; 11:biom11030464. [PMID: 33804786 PMCID: PMC8035981 DOI: 10.3390/biom11030464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Telomeres are repetitive nucleoprotein DNA sequences that shorten with each cell division. The stem cells activate telomerase to compensate for the telomere loss. This study aimed to evaluate the effect of cultivation passaging on the relative telomere length and proliferation capacity of dental pulp stem cells. We used ten dental pulp stem cell (DPSC) lineages stored for 12 months using uncontrolled-rate freezing to reach the study’s goal. We analyzed their proliferation rate, phenotype using flow cytometry, multipotency, and relative telomere length using a qPCR analysis. We determined the relative telomere length in the added study by performing analysis after one, two, and three weeks of cultivation with no passaging. We documented the telomere attrition with increasing passaging. The shorter the relative telomere length, the lower reached population doublings, and longer population doubling time were observed at the end of the cultivation. We observed the telomere prolongation in DPSCs cultivated for two weeks with no passaging in the added subsequent study. We concluded that excessive proliferation demands on DPSCs during in vitro cultivation result in telomere attrition. We opened the theory that the telomerase might be more efficient during cell cultivation with no passaging. This observation could help in preserving the telomere length during ex vivo DPSC expansion.
Collapse
|
10
|
Xie W, Zhu H, Zhao M, Wang L, Li S, Zhao C, Zhou Y, Zhu B, Jiang X, Liu W, Ren C. Crucial roles of different RNA-binding hnRNP proteins in Stem Cells. Int J Biol Sci 2021; 17:807-817. [PMID: 33767590 PMCID: PMC7975692 DOI: 10.7150/ijbs.55120] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/07/2021] [Indexed: 11/05/2022] Open
Abstract
The self-renewal, pluripotency and differentiation of stem cells are regulated by various genetic and epigenetic factors. As a kind of RNA binding protein (RBP), the heterogeneous nuclear ribonucleoproteins (hnRNPs) can act as "RNA scaffold" and recruit mRNA, lncRNA, microRNA and circRNA to affect mRNA splicing and processing, regulate gene transcription and post-transcriptional translation, change genome structure, and ultimately play crucial roles in the biological processes of cells. Recent researches have demonstrated that hnRNPs are irreplaceable for self-renewal and differentiation of stem cells. hnRNPs function in stem cells by multiple mechanisms, which include regulating mRNA stability, inducing alternative splicing of mRNA, epigenetically regulate gene expression, and maintaining telomerase activity and telomere length. The functions and the underlying mechanisms of hnRNPs in stem cells deserve further investigation.
Collapse
Affiliation(s)
- Wen Xie
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Hecheng Zhu
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,Changsha Kexin Cancer Hospital, Changsha, Hunan 410205, China
| | - Ming Zhao
- Changsha Kexin Cancer Hospital, Changsha, Hunan 410205, China
| | - Lei Wang
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Shasha Li
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Cong Zhao
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Yao Zhou
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Bin Zhu
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Xingjun Jiang
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Weidong Liu
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| | - Caiping Ren
- Cancer Research Institute, Department of Neurosurgery, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha 410008, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha 410008, China
| |
Collapse
|
11
|
Carlone DL, Riba-Wolman RD, Deary LT, Tovaglieri A, Jiang L, Ambruzs DM, Mead BE, Shah MS, Lengner CJ, Jaenisch R, Breault DT. Telomerase expression marks transitional growth-associated skeletal progenitor/stem cells. Stem Cells 2021; 39:296-305. [PMID: 33438789 PMCID: PMC7986156 DOI: 10.1002/stem.3318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Skeletal progenitor/stem cells (SSCs) play a critical role in postnatal bone growth and maintenance. Telomerase (Tert) activity prevents cellular senescence and is required for maintenance of stem cells in self‐renewing tissues. Here we investigated the role of mTert‐expressing cells in postnatal mouse long bone and found that mTert expression is enriched at the time of adolescent bone growth. mTert‐GFP+ cells were identified in regions known to house SSCs, including the metaphyseal stroma, growth plate, and the bone marrow. We also show that mTert‐expressing cells are a distinct SSC population with enriched colony‐forming capacity and contribute to multiple mesenchymal lineages, in vitro. In contrast, in vivo lineage‐tracing studies identified mTert+ cells as osteochondral progenitors and contribute to the bone‐forming cell pool during endochondral bone growth with a subset persisting into adulthood. Taken together, our results show that mTert expression is temporally regulated and marks SSCs during a discrete phase of transitional growth between rapid bone growth and maintenance.
Collapse
Affiliation(s)
- Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Rebecca D Riba-Wolman
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Luke T Deary
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alessio Tovaglieri
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lijie Jiang
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Dana M Ambruzs
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Benjamin E Mead
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Manasvi S Shah
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
12
|
Li H, Wang B, Li D, Li J, Luo Y, Dan J. Roles of telomeres and telomerase in age‑related renal diseases (Review). Mol Med Rep 2020; 23:96. [PMID: 33300081 PMCID: PMC7723152 DOI: 10.3892/mmr.2020.11735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/30/2020] [Indexed: 01/20/2023] Open
Abstract
Age‑related renal diseases, which account for various progressive renal disorders associated with cellular and organismal senescence, are becoming a substantial public health burden. However, their aetiologies are complicated and their pathogeneses remain poorly understood. Telomeres and telomerase are known to be essential for maintaining the integrity and stability of eukaryotic genomes and serve crucial roles in numerous related signalling pathways that activate renal functions, such as repair and regeneration. Previous studies have reported that telomere dysfunction served a role in various types of age‑related kidney disease through various different molecular pathways. The present review aimed to summarise the current knowledge of the association between telomeres and ageing‑related kidney diseases and explored the contribution of dysfunctional telomeres to these diseases. The findings may help to provide novel strategies for treating patients with renal disease.
Collapse
Affiliation(s)
- Haili Li
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Boyuan Wang
- The Key Lab of Sports and Rehabilitation, Faculty of Physical Education, Yuxi Normal University, Yuxi, Yunnan 653100, P.R. China
| | - Daoqun Li
- Department of Human Anatomy, School of Basic Medicine and Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong 250014, P.R. China
| | - Jinyuan Li
- Department of General Surgery, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Ying Luo
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
13
|
Wu L, Fidan K, Um JY, Ahn KS. Telomerase: Key regulator of inflammation and cancer. Pharmacol Res 2020; 155:104726. [PMID: 32109579 DOI: 10.1016/j.phrs.2020.104726] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
The telomerase holoenzyme, which has a highly conserved role in maintaining telomere length, has long been regarded as a high-profile target in cancer therapy due to the high dependency of the majority of cancer cells on constitutive and elevated telomerase activity for sustained proliferation and immortality. In this review, we present the salient findings in the telomerase field with special focus on the association of telomerase with inflammation and cancer. The elucidation of extra-telomeric roles of telomerase in inflammation, reactive oxygen species (ROS) generation, and cancer development further complicated the design of anti-telomerase therapy. Of note, the discovery of the unique mechanism that underlies reactivation of the dormant telomerase reverse transcriptase TERT promoter in somatic cells not only enhanced our understanding of the critical role of TERT in carcinogenesis but also opens up new intervention ideas that enable the differential targeting of cancer cells only. Despite significant effort invested in developing telomerase-targeted therapeutics, devising efficacious cancer-specific telomerase/TERT inhibitors remains an uphill task. The latest discoveries of the telomere-independent functionalities of telomerase in inflammation and cancer can help illuminate the path of developing specific anti-telomerase/TERT therapeutics against cancer cells.
Collapse
Affiliation(s)
- Lele Wu
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore
| | - Kerem Fidan
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Jae-Young Um
- College of Korean Medicine, Kyung Hee University, #47, Kyungheedae-gil, Dongdaemoon-gu, Seoul 130-701, Republic of Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, #47, Kyungheedae-gil, Dongdaemoon-gu, Seoul 130-701, Republic of Korea.
| |
Collapse
|
14
|
Wang Z, Li S, Wang Y, Zhang X, Chen L, Sun D. GDNF enhances the anti-inflammatory effect of human adipose-derived mesenchymal stem cell-based therapy in renal interstitial fibrosis. Stem Cell Res 2019; 41:101605. [PMID: 31706095 DOI: 10.1016/j.scr.2019.101605] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (AMSCs) are a type of adult stem cell from the mesoderm with the capacity to migrate and differentiate into other cell lineages. As a morphogenetic state of stem cells, glial-derived neurotrophic factor (GDNF) has been found to promote cell proliferation and differentiation of stem cells. The aims of our study were to investigate the biological activity of AMSCs and whether the GDNF gene can enhance the anti-inflammatory properties of stem cells. In this study, stable proliferative GDNF-overexpressing AMSC lines were successfully established and the AMSCs/GDNF-AMSCs were cocultured with macrophages (Mφ) derived from THP-1 cells in a transwell system. The mRNA expression levels of tumor necrosis factor-alpha (TNF-α), inducible nitric oxide synthase (iNOS), interleukin (IL)-10 and IL-4 were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). In addition, the expressions of CD163 and CD206, two markers of M2 macrophages, were detected with flow cytometric analysis. In animal experiments, AMSCs/GDNF-AMSCs (5 × 105) were administered to unilateral ureteral obstruction (UUO) nude mice for 3 or 7 days. The expression levels of cyclooxygenase-2 (COX-2), IL-6, transforming growth factor β1 (TGF-β1) and α-Smooth muscle actin (α-SMA) were determined by Western blotting. Renal pathological changes of all groups were observed by hematoxylin and eosin (HE) and Masson staining. In conclusion, in vitro cultured AMSCs induced a shift in macrophage phenotype from the inflammatory (M1) phenotype to the reparative (M2) phenotype. In the UUO model, AMSC treatment was conducive to the recovery of renal function and interstitial fibrosis. Therefore, we determined that AMSC therapy could promote the phenotypic transformation of macrophages and reduce the progression of renal fibrosis by suppressing inflammation. GDNF could enhance the anti-inflammatory effect of AMSCs.
Collapse
Affiliation(s)
- Zhuojun Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Yanping Wang
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou 221002, China
| | - Xiangyu Zhang
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou 221002, China
| | - Lu Chen
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou 221002, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
15
|
Increased Gene Expression of RUNX2 and SOX9 in Mesenchymal Circulating Progenitors Is Associated with Autophagy during Physical Activity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8426259. [PMID: 31737174 PMCID: PMC6815530 DOI: 10.1155/2019/8426259] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/22/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Lack of physical exercise is considered an important risk factor for chronic diseases. On the contrary, physical exercise reduces the morbidity rates of obesity, diabetes, bone disease, and hypertension. In order to gain novel molecular and cellular clues, we analyzed the effects of physical exercise on differentiation of mesenchymal circulating progenitor cells (M-CPCs) obtained from runners. We also investigated autophagy and telomerase-related gene expression to evaluate the involvement of specific cellular functions in the differentiation process. We performed cellular and molecular analyses in M-CPCs, obtained by a depletion method, of 22 subjects before (PRE RUN) and after (POST RUN) a half marathon performance. In order to prove our findings, we performed also in vitro analyses by testing the effects of runners' sera on a human bone marrow-derived mesenchymal stem (hBM-MSC) cell line. PCR array analyses of PRE RUN versus POST RUN M-CPC total RNAs put in evidence several genes which appeared to be modulated by physical activity. Our results showed that physical exercise promotes differentiation. Osteogenesis-related genes as RUNX2, MSX1, and SPP1 appeared to be upregulated after the run; data showed also increased levels of BMP2 and BMP6 expressions. SOX9, COL2A1, and COMP gene enhanced expression suggested the induction of chondrocytic differentiation as well. The expression of telomerase-associated genes and of two autophagy-related genes, ATG3 and ULK1, was also affected and correlated positively with MSC differentiation. These data highlight an attractive cellular scenario, outlining the role of autophagic response to physical exercise and suggesting new insights into the benefits of physical exercise in counteracting chronic degenerative conditions.
Collapse
|
16
|
Base Editor Correction of COL7A1 in Recessive Dystrophic Epidermolysis Bullosa Patient-Derived Fibroblasts and iPSCs. THE JOURNAL OF INVESTIGATIVE DERMATOLOGY 2019. [PMID: 31437443 DOI: 10.1016/j.jid.2019.07.701.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Genome editing represents a promising strategy for the therapeutic correction of COL7A1 mutations that cause recessive dystrophic epidermolysis bullosa (RDEB). DNA cleavage followed by homology-directed repair (HDR) using an exogenous template has previously been used to correct COL7A1 mutations. HDR rates can be modest, and the double-strand DNA breaks that initiate HDR commonly result in accompanying undesired insertions and deletions (indels). To overcome these limitations, we applied an A•T→G•C adenine base editor (ABE) to correct two different COL7A1 mutations in primary fibroblasts derived from RDEB patients. ABE enabled higher COL7A1 correction efficiencies than previously reported HDR efforts. Moreover, ABE obviated the need for a repair template, and minimal indels or editing at off-target sites was detected. Base editing restored the endogenous type VII collagen expression and function in vitro. We also treated induced pluripotent stem cells (iPSCs) derived from RDEB fibroblasts with ABE. The edited iPSCs were differentiated into mesenchymal stromal cells, a cell population with therapeutic potential for RDEB. In a mouse teratoma model, the skin derived from ABE-treated iPSCs showed the proper deposition of C7 at the dermal-epidermal junction in vivo. These demonstrate that base editing provides an efficient and precise genome editing method for autologous cell engineering for RDEB.
Collapse
|
17
|
Base Editor Correction of COL7A1 in Recessive Dystrophic Epidermolysis Bullosa Patient-Derived Fibroblasts and iPSCs. J Invest Dermatol 2019; 140:338-347.e5. [PMID: 31437443 DOI: 10.1016/j.jid.2019.07.701] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/18/2019] [Accepted: 07/29/2019] [Indexed: 12/14/2022]
Abstract
Genome editing represents a promising strategy for the therapeutic correction of COL7A1 mutations that cause recessive dystrophic epidermolysis bullosa (RDEB). DNA cleavage followed by homology-directed repair (HDR) using an exogenous template has previously been used to correct COL7A1 mutations. HDR rates can be modest, and the double-strand DNA breaks that initiate HDR commonly result in accompanying undesired insertions and deletions (indels). To overcome these limitations, we applied an A•T→G•C adenine base editor (ABE) to correct two different COL7A1 mutations in primary fibroblasts derived from RDEB patients. ABE enabled higher COL7A1 correction efficiencies than previously reported HDR efforts. Moreover, ABE obviated the need for a repair template, and minimal indels or editing at off-target sites was detected. Base editing restored the endogenous type VII collagen expression and function in vitro. We also treated induced pluripotent stem cells (iPSCs) derived from RDEB fibroblasts with ABE. The edited iPSCs were differentiated into mesenchymal stromal cells, a cell population with therapeutic potential for RDEB. In a mouse teratoma model, the skin derived from ABE-treated iPSCs showed the proper deposition of C7 at the dermal-epidermal junction in vivo. These demonstrate that base editing provides an efficient and precise genome editing method for autologous cell engineering for RDEB.
Collapse
|
18
|
Li J, Liu W, Yao W. Immortalized Human Bone Marrow Derived Stromal Cells in Treatment of Transient Cerebral Ischemia in Rats. J Alzheimers Dis 2019; 69:871-880. [PMID: 31156178 DOI: 10.3233/jad-190279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jianyuan Li
- Neurosurgical Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Neurosurgical Department, Rizhao City Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Weidong Liu
- Neurosurgical Department, Liaocheng People’s Hospital, Liaocheng, China
| | - Weicheng Yao
- Neurosurgical Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Fitzsimmons REB, Mazurek MS, Soos A, Simmons CA. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem Cells Int 2018; 2018:8031718. [PMID: 30210552 PMCID: PMC6120267 DOI: 10.1155/2018/8031718] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/08/2023] Open
Abstract
As a result of over five decades of investigation, mesenchymal stromal/stem cells (MSCs) have emerged as a versatile and frequently utilized cell source in the fields of regenerative medicine and tissue engineering. In this review, we summarize the history of MSC research from the initial discovery of their multipotency to the more recent recognition of their perivascular identity in vivo and their extraordinary capacity for immunomodulation and angiogenic signaling. As well, we discuss long-standing questions regarding their developmental origins and their capacity for differentiation toward a range of cell lineages. We also highlight important considerations and potential risks involved with their isolation, ex vivo expansion, and clinical use. Overall, this review aims to serve as an overview of the breadth of research that has demonstrated the utility of MSCs in a wide range of clinical contexts and continues to unravel the mechanisms by which these cells exert their therapeutic effects.
Collapse
Affiliation(s)
- Ross E. B. Fitzsimmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Matthew S. Mazurek
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada T2N 4Z6
| | - Agnes Soos
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Craig A. Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada M5S 3G8
| |
Collapse
|
20
|
Liu MY, Nemes A, Zhou QG. The Emerging Roles for Telomerase in the Central Nervous System. Front Mol Neurosci 2018; 11:160. [PMID: 29867352 PMCID: PMC5964194 DOI: 10.3389/fnmol.2018.00160] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Telomerase, a specialized ribonucleoprotein enzyme complex, maintains telomere length at the 3′ end of chromosomes, and functions importantly in stem cells, cancer and aging. Telomerase exists in neural stem cells (NSCs) and neural progenitor cells (NPCs), at a high level in the developing and adult brains of humans and rodents. Increasing studies have demonstrated that telomerase in NSCs/NPCs plays important roles in cell proliferation, neuronal differentiation, neuronal survival and neuritogenesis. In addition, recent works have shown that telomerase reverse transcriptase (TERT) can protect newborn neurons from apoptosis and excitotoxicity. However, to date, the link between telomerase and diseases in the central nervous system (CNS) is not well reviewed. Here, we analyze the evidence and summarize the important roles of telomerase in the CNS. Understanding the roles of telomerase in the nervous system is not only important to gain further insight into the process of the neural cell life cycle but would also provide novel therapeutic applications in CNS diseases such as neurodegenerative condition, mood disorders, aging and other ailments.
Collapse
Affiliation(s)
- Meng-Ying Liu
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China.,The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, China
| | - Ashley Nemes
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China.,Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
21
|
El Agha E, Kramann R, Schneider RK, Li X, Seeger W, Humphreys BD, Bellusci S. Mesenchymal Stem Cells in Fibrotic Disease. Cell Stem Cell 2018; 21:166-177. [PMID: 28777943 DOI: 10.1016/j.stem.2017.07.011] [Citation(s) in RCA: 300] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibrosis is associated with organ failure and high mortality and is commonly characterized by aberrant myofibroblast accumulation. Investigating the cellular origin of myofibroblasts in various diseases is thus a promising strategy for developing targeted anti-fibrotic treatments. Recent studies using genetic lineage tracing technology have implicated diverse organ-resident perivascular mesenchymal stem cell (MSC)-like cells and bone marrow-MSCs in myofibroblast generation during fibrosis development. In this Review, we give an overview of the emerging role of MSCs and MSC-like cells in myofibroblast-mediated fibrotic disease in the kidney, lung, heart, liver, skin, and bone marrow.
Collapse
Affiliation(s)
- Elie El Agha
- Institute of Life Sciences, Wenzhou University, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedicine, Wenzhou, Zhejiang, China; Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany.
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, Medical Faculty RWTH Aachen University, RWTH Aachen University, Aachen, Germany
| | - Rebekka K Schneider
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Xiaokun Li
- Institute of Life Sciences, Wenzhou University, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedicine, Wenzhou, Zhejiang, China
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany; Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, Bad Nauheim, Germany
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, USA
| | - Saverio Bellusci
- Institute of Life Sciences, Wenzhou University, Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedicine, Wenzhou, Zhejiang, China; Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
22
|
Role of Mesenchymal Stem Cells in Cancer Development and Their Use in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:45-62. [DOI: 10.1007/5584_2017_64] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Variation in human dental pulp stem cell ageing profiles reflect contrasting proliferative and regenerative capabilities. BMC Cell Biol 2017; 18:12. [PMID: 28148303 PMCID: PMC5288874 DOI: 10.1186/s12860-017-0128-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 01/20/2017] [Indexed: 12/17/2022] Open
Abstract
Background Dental pulp stem cells (DPSCs) are increasingly being recognized as a viable cell source for regenerative medicine. Although significant variations in their ex vivo expansion are well-established, DPSC proliferative heterogeneity remains poorly understood, despite such characteristics influencing their regenerative and therapeutic potential. This study assessed clonal human DPSC regenerative potential and the impact of cellular senescence on these responses, to better understand DPSC functional behaviour. Results All DPSCs were negative for hTERT. Whilst one DPSC population reached >80 PDs before senescence, other populations only achieved <40 PDs, correlating with DPSCs with high proliferative capacities possessing longer telomeres (18.9 kb) than less proliferative populations (5–13 kb). High proliferative capacity DPSCs exhibited prolonged stem cell marker expression, but lacked CD271. Early-onset senescence, stem cell marker loss and positive CD271 expression in DPSCs with low proliferative capacities were associated with impaired osteogenic and chondrogenic differentiation, favouring adipogenesis. DPSCs with high proliferative capacities only demonstrated impaired differentiation following prolonged expansion (>60 PDs). Conclusions This study has identified that proliferative and regenerative heterogeneity is related to contrasting telomere lengths and CD271 expression between DPSC populations. These characteristics may ultimately be used to selectively screen and isolate high proliferative capacity/multi-potent DPSCs for regenerative medicine exploitation.
Collapse
|
24
|
Ma C, Pi C, Yang Y, Lin L, Shi Y, Li Y, Li Y, He X. Nampt Expression Decreases Age-Related Senescence in Rat Bone Marrow Mesenchymal Stem Cells by Targeting Sirt1. PLoS One 2017; 12:e0170930. [PMID: 28125705 PMCID: PMC5268649 DOI: 10.1371/journal.pone.0170930] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
Senescence restricts the development of applications involving mesenchymal stem cells (MSCs) in research fields, such as tissue engineering, and stem cell therapeutic strategies. Understanding the mechanisms underlying natural aging processes may contribute to the development of novel approaches to preventing age-related diseases or slowing individual aging processes. Nampt is a rate-limiting NAD biosynthetic enzyme that plays critical roles in energy metabolism, cell senescence and maintaining life spans. However, it remains unknown whether Nampt influences stem cell senescence. In this study, the function of Nampt was investigated using a rat model of natural aging. Our data show that Nampt expression was significantly lower in MSCs obtained from aged rats than in those obtained from young rats during physiological aging. Reducing the level of Nampt in aged MSCs resulted in lower intracellular concentrations of NAD+ and downregulated Sirt1 expression and activity. After the Nampt inhibitor FK866 was added, young MSCs were induced to become aged cells. The enhanced senescence was correlated with NAD+ depletion and Sirt1 activity attenuation. In addition, Nampt overexpression attenuated cell senescence in aged MSCs. Our findings provide a new explanation for the mechanisms underlying stem cell senescence and a novel target for delaying stem cell senescence and preventing and treating age-related diseases.
Collapse
Affiliation(s)
- Cao Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chenchen Pi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yue Yang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lin Lin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yan Li
- Center for Advanced Reconstruction of Extremities (C.A.R.E.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
25
|
Dzobo K, Turnley T, Wishart A, Rowe A, Kallmeyer K, van Vollenstee FA, Thomford NE, Dandara C, Chopera D, Pepper MS, Parker MI. Fibroblast-Derived Extracellular Matrix Induces Chondrogenic Differentiation in Human Adipose-Derived Mesenchymal Stromal/Stem Cells in Vitro. Int J Mol Sci 2016; 17:E1259. [PMID: 27527147 PMCID: PMC5000657 DOI: 10.3390/ijms17081259] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 07/17/2016] [Accepted: 07/25/2016] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) represent an area being intensively researched for tissue engineering and regenerative medicine applications. MSCs may provide the opportunity to treat diseases and injuries that currently have limited therapeutic options, as well as enhance present strategies for tissue repair. The cellular environment has a significant role in cellular development and differentiation through cell-matrix interactions. The aim of this study was to investigate the behavior of adipose-derived MSCs (ad-MSCs) in the context of a cell-derived matrix so as to model the in vivo physiological microenvironment. The fibroblast-derived extracellular matrix (fd-ECM) did not affect ad-MSC morphology, but reduced ad-MSC proliferation. Ad-MSCs cultured on fd-ECM displayed decreased expression of integrins α2 and β1 and subsequently lost their multipotency over time, as shown by the decrease in CD44, Octamer-binding transcription factor 4 (OCT4), SOX2, and NANOG gene expression. The fd-ECM induced chondrogenic differentiation in ad-MSCs compared to control ad-MSCs. Loss of function studies, through the use of siRNA and a mutant Notch1 construct, revealed that ECM-mediated ad-MSCs chondrogenesis requires Notch1 and β-catenin signaling. The fd-ECM also showed anti-senescence effects on ad-MSCs. The fd-ECM is a promising approach for inducing chondrogenesis in ad-MSCs and chondrogenic differentiated ad-MSCs could be used in stem cell therapy procedures.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Anzio Road, Observatory, Cape Town 7925, South Africa.
- Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Taegyn Turnley
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Anzio Road, Observatory, Cape Town 7925, South Africa.
- Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Andrew Wishart
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Anzio Road, Observatory, Cape Town 7925, South Africa.
- Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Arielle Rowe
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Karlien Kallmeyer
- Department of Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa.
| | - Fiona A van Vollenstee
- Department of Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa.
| | - Nicholas E Thomford
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Denis Chopera
- Division of Immunology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Michael S Pepper
- Department of Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa.
| | - M Iqbal Parker
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Anzio Road, Observatory, Cape Town 7925, South Africa.
- Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| |
Collapse
|
26
|
Deane JA, Ong YR, Cain JE, Jayasekara WSN, Tiwari A, Carlone DL, Watkins DN, Breault DT, Gargett CE. The mouse endometrium contains epithelial, endothelial and leucocyte populations expressing the stem cell marker telomerase reverse transcriptase. Mol Hum Reprod 2016; 22:272-84. [PMID: 26740067 DOI: 10.1093/molehr/gav076] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/22/2015] [Indexed: 12/11/2022] Open
Abstract
STUDY HYPOTHESIS The mouse endometrium harbours stem/progenitor cells that express the stem cell marker mouse telomerase reverse transcriptase (mTert). STUDY FINDING We used a mouse carrying a transgenic reporter for mTert promoter activity to identify rare endometrial populations of epithelial and endothelial cells that express mTert. WHAT IS KNOWN ALREADY Stem/progenitor cells are hypothesized to be responsible for the remarkable regenerative capacity of the endometrium, but the lack of convenient endometrial stem/progenitor markers in the mouse has hampered investigations into the identity of these cells. STUDY DESIGN, SAMPLES/MATERIALS, METHODS A mouse containing a green fluorescent protein (GFP) reporter under the control of the telomerase reverse transcriptase promoter (mTert-GFP) was used to identify potential stem/progenitor cells in the endometrium. mTert promoter activity was determined using fluorescence microscopy and flow cytometry to identify GFP(+) cells. GFP(+) cells were examined for epithelial, stromal, endothelial, leucocyte and proliferation markers and bromodeoxyuridine retention to determine their identity. The endometrium of ovariectomized mice was compared to that of intact cycling mice to establish the role of ovarian hormones in maintaining mTert-expressing cells. MAIN RESULTS AND THE ROLE OF CHANCE We found that mTert-GFP is expressed by rare luminal and glandular epithelial cells (0.3% of epithelial cells by flow cytometry), rare CD45(-) cells in the stromal compartment (0.028 ± 0.010% of stromal cells by microscopy) and many CD45(+) leucocytes. Ovariectomy resulted in significant decrease of mTert-GFP(+) epithelial cells (P = 0.029 for luminal epithelium; P = 0.034 for glandular epithelium) and a decrease in the percentage of mTert-GFP(+) CD45(+) leucocytes in the stromal compartment (P = 0.015). However, CD45(-) mTert-GFP(+) cells in the stromal compartment were maintained in ovariectomized mice. This population is enriched for cells bearing the endothelial marker CD31 (10.3% of CD90(-) CD45(-) and 97.8% CD90(+) CD45(-) by flow cytometry). CD45(-) mTert-GFP(+) cells also immunostained for the endothelial marker von Willebrand factor. These results suggest that the endometrial epithelium and vasculature are foci of stem/progenitor activity and provide a system to investigate molecular mechanisms involved in endometrial regeneration and repair. LIMITATIONS, REASONS FOR CAUTION The stem/progenitor activity of endometrial mTert-GFP(+) cells needs to be experimentally verified. WIDER IMPLICATIONS OF THE FINDINGS The identification and characterization of mTert-expressing progenitor cells in the mouse will facilitate the identification of equivalent populations in the human endometrium that are likely to be involved in endometrial function, fertility and disease. LARGE-SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This study was funded by National Health and Medical Research Council (NHMRC) of Australia grants (1085435, C.E.G., J.A.D.), 1021127 (C.E.G.), NHMRC Senior Research Fellowship (1042298, C.E.G.), the Victorian Infrastructure Support Program, U.S. National Institutes of Health grant R01 DK084056 (D.T.B.) and the Harvard Stem Cell Institute (D.T.B.). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- James A Deane
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Y Rue Ong
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia
| | - Jason E Cain
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - W Samantha N Jayasekara
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Abhilasha Tiwari
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia
| | - Diana L Carlone
- Boston Children's Hospital, Harvard Medical School/Harvard Stem Cell Institute, Boston, MA, USA
| | - D Neil Watkins
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia Present address: The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia Present address: UNSW Faculty of Medicine, St Vincent's Clinical School, St Vincent's Hospital, Randwick, New South Wales, Australia
| | - David T Breault
- Boston Children's Hospital, Harvard Medical School/Harvard Stem Cell Institute, Boston, MA, USA
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
27
|
Jeon BG, Bharti D, Lee WJ, Jang SJ, Park JS, Jeong GJ, Rho GJ. Comparison of mesenchymal stem cells isolated from various tissues of isogenic mini-pig. Anim Cells Syst (Seoul) 2015. [DOI: 10.1080/19768354.2015.1089323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
28
|
Trivanović D, Jauković A, Popović B, Krstić J, Mojsilović S, Okić-Djordjević I, Kukolj T, Obradović H, Santibanez JF, Bugarski D. Mesenchymal stem cells of different origin: Comparative evaluation of proliferative capacity, telomere length and pluripotency marker expression. Life Sci 2015; 141:61-73. [PMID: 26408916 DOI: 10.1016/j.lfs.2015.09.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 08/05/2015] [Accepted: 09/22/2015] [Indexed: 01/07/2023]
Abstract
AIMS In vitro expansion changes replication and differentiation capacity of mesenchymal stem cells (MSCs), increasing challenges and risks, while limiting the sufficient number of MSCs required for cytotherapy. Here, we characterized and compared proliferation, differentiation, telomere length and pluripotency marker expression in MSCs of various origins. MAIN METHODS Immunophenotyping, proliferation and differentiation assays were performed. Pluripotency marker (Nanog, Oct-4, SOX-2, SSEA-4) expression was determined by immunofluorescence. Quantitative PCR was performed for relative telomere length (RTL) analyses, while expression of relevant genes for pluripotency markers, differentiation state (Cbfa1, human placental alkaline phosphatase, peroxisome proliferator activated receptor, Sox9 and Collagen II a1), and telomerase reverse transcriptase (hTERT) was determined by semiquantitative RT-PCR. KEY FINDINGS Peripheral blood MSCs (PB-MSCs) and umbilical cord MSCs (UC-MSCs) showed the highest, while periodontal ligament MSCs (PDL-MSCs) and adipose tissue MSCs (AT-MSCs) the lowest values of both the replication potential and RTL. Although MSCs from exfoliated deciduous teeth (SHEDs), PDL-MSCs and AT-MSCs showed higher mRNA expression of pluripotency markers, all MSCs expressed pluripotency marker proteins. SHEDs and PDL-MSCs showed prominent capacity for osteogenesis, PB-MSCs and UC-MSCs showed strengthened adipogenic differentiation potential, while AT-MSCs displayed similar differentiation into both lines. SIGNIFICANCE The MSCs populations derived from different sources, although displaying similar phenotype, exhibited high degree of variability regarding biological properties related to their self-renewal and differentiation capacity. These data indicate that for more accurate use in cell therapy, individualities of MSCs isolated from different tissues should be identified and taken into consideration when planning their use in clinical protocols.
Collapse
Affiliation(s)
- Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Branka Popović
- Institute of Human Genetics, School of Dentistry, University of Belgrade, Belgrade, Serbia
| | - Jelena Krstić
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Slavko Mojsilović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Ivana Okić-Djordjević
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Hristina Obradović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Juan Francisco Santibanez
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
29
|
Sui B, Hu C, Jin Y. Mitochondrial metabolic failure in telomere attrition-provoked aging of bone marrow mesenchymal stem cells. Biogerontology 2015; 17:267-79. [DOI: 10.1007/s10522-015-9609-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/15/2015] [Indexed: 12/16/2022]
|
30
|
Ma D, Kua JEH, Lim WK, Lee ST, Chua AWC. In vitro characterization of human hair follicle dermal sheath mesenchymal stromal cells and their potential in enhancing diabetic wound healing. Cytotherapy 2015; 17:1036-51. [DOI: 10.1016/j.jcyt.2015.04.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 01/09/2023]
|
31
|
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: 0.9] [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
|
32
|
ZHAO QIANG, WANG XUEYAO, YU XIAOXIA, ZHAI YINGXIAN, HE XU, WU SHAN, SHI YINGAI. Expression of human telomerase reverse transcriptase mediates the senescence of mesenchymal stem cells through the PI3K/AKT signaling pathway. Int J Mol Med 2015; 36:857-64. [DOI: 10.3892/ijmm.2015.2284] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/08/2015] [Indexed: 11/06/2022] Open
|
33
|
Fu WL, Li J, Chen G, Li Q, Tang X, Zhang CH. Mesenchymal Stem Cells Derived from Peripheral Blood Retain Their Pluripotency, but Undergo Senescence During Long-Term Culture. Tissue Eng Part C Methods 2015; 21:1088-97. [PMID: 25996678 DOI: 10.1089/ten.tec.2014.0595] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Peripheral blood-derived mesenchymal stem cells (PB-MSCs) show promise as a source of cells for autologous transplantation because they can be harvested through minimally invasive procedures. To ensure adequate numbers of cells for transplantation and tissue regeneration, PB-MSCs must first be cultured and expanded in vitro, but whether long-term passage modifies their properties has been poorly understood. In this study we triggered production of PB-MSCs in rabbits using granulocyte colony-stimulating factor (G-CSF) and AMD3100, and then isolated and expanded the cells in culture until they reached a state of senescence, usually after about 20 passages. Cultures of low-, middle-, and high-passage numbers were compared in terms of morphology, proliferative capacity, phenotype, differentiation potential, apoptosis, metabolic indicators, and senescence. As passage number increased, MSCs retained their elongated spindle shape, but became larger and flatter, slowed in growth gradually, and increased proportion of cells showed G1 arrest. The proportions of apoptotic cells, production of reactive oxygen species (ROS), and ADP/ATP ratio increased with passage number. Expression of senescence-associated β-galactosidase increased, while telomerase activity decreased. On the other hand, cultures did not show significant changes in phenotype or lose their ability to differentiate into three lineages as passage number increased. These results suggest that PB-MSCs maintain their stem cell properties during prolonged culturing, but they undergo senescence that may be due to apoptosis and production of ROS. These findings may help to standardize in vitro production of PB-MSCs for tissue engineering.
Collapse
Affiliation(s)
- Wei-Li Fu
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Jian Li
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Gang Chen
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Qi Li
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Xin Tang
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| | - Cheng-Hao Zhang
- Department of Orthopaedics, West China Hospital, Sichuan University , Chengdu, P.R. China
| |
Collapse
|
34
|
Chen H, Liu X, Zhu W, Chen H, Hu X, Jiang Z, Xu Y, Wang L, Zhou Y, Chen P, Zhang N, Hu D, Zhang L, Wang Y, Xu Q, Wu R, Yu H, Wang J. SIRT1 ameliorates age-related senescence of mesenchymal stem cells via modulating telomere shelterin. Front Aging Neurosci 2014; 6:103. [PMID: 24917814 PMCID: PMC4042159 DOI: 10.3389/fnagi.2014.00103] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/14/2014] [Indexed: 01/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs) senescence is an age-related process that impairs the capacity for tissue repair and compromises the clinical use of autologous MSCs for tissue regeneration. Here, we describe the effects of SIRT1, a NAD+-dependent deacetylase, on age-related MSCs senescence. Knockdown of SIRT1 in young MSCs induced cellular senescence and inhibited cell proliferation whereas overexpression of SIRT1 in aged MSCs reversed the senescence phenotype and stimulated cell proliferation. These results suggest that SIRT1 plays a key role in modulating age-induced MSCs senescence. Aging-related proteins, P16 and P21 may be downstream effectors of the SIRT1-mediated anti-aging effects. SIRT1 protected MSCs from age-related DNA damage, induced telomerase reverse transcriptase (TERT) expression and enhanced telomerase activity but did not affect telomere length. SIRT1 positively regulated the expression of tripeptidyl peptidase 1 (TPP1), a component of the shelterin pathway that protects chromosome ends from DNA damage. Together, the results demonstrate that SIRT1 quenches age-related MSCs senescence by mechanisms that include enhanced TPP1 expression, increased telomerase activity and reduced DNA damage.
Collapse
Affiliation(s)
- Huiqiang Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Xianbao Liu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Wei Zhu
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Han Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Zhi Jiang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Yinchuan Xu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Lihan Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Yu Zhou
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Panpan Chen
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Na Zhang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Dexing Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Ling Zhang
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Yaping Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Qiyuan Xu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Rongrong Wu
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Hong Yu
- Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China ; Key Lab of Cardiovascular Disease, Second Affiliated Hospital, College of Medicine, Zhejiang University Hangzhou, China
| |
Collapse
|
35
|
Salehinejad P, Alitheen NB, Mandegary A, Nematollahi-mahani SN, Janzamin E. Effect of EGF and FGF on the expansion properties of human umbilical cord mesenchymal cells. In Vitro Cell Dev Biol Anim 2013; 49:515-23. [DOI: 10.1007/s11626-013-9631-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 04/29/2013] [Indexed: 12/13/2022]
|
36
|
Smith LB, Belanger JM, Oberbauer AM. Fibroblast growth factor receptor 3 effects on proliferation and telomerase activity in sheep growth plate chondrocytes. J Anim Sci Biotechnol 2012; 3:39. [PMID: 23216972 PMCID: PMC3541258 DOI: 10.1186/2049-1891-3-39] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 12/03/2012] [Indexed: 11/10/2022] Open
Abstract
UNLABELLED BACKGROUND Fibroblast growth factor receptor 3 (FGFR3) inhibits growth-plate chondrocyte proliferation and limits bone elongation. Gain-of-function FGFR3 mutations cause dwarfism, reduced telomerase activity and shorter telomeres in growth plate chondroyctes suggesting that FGFR3 reduces proliferative capacity, inhibits telomerase, and enhances senescence. Thyroid hormone (T3) plays a role in cellular maturation of growth plate chondrocytes and a known target of T3 is FGFR3. The present study addressed whether reduced FGFR3 expression enhanced telomerase activity, mRNA expression of telomerase reverse transcriptase (TERT) and RNA component of telomerase (TR), and chondrocyte proliferation, and whether the stimulation of FGFR3 by T3 evoked the opposite response. RESULTS Sheep growth-plate proliferative zone chondrocytes were cultured and transfected with siRNA to reduce FGFR3 expression; FGFR3 siRNA reduced chondrocyte FGFR3 mRNA and protein resulting in greater proliferation and increased TERT mRNA expression and telomerase activity (p < 0.05). Chondrocytes treated with T3 significantly enhanced FGFR3 mRNA and protein expression and reduced telomerase activity (p < 0.05); TERT and TR were not significantly reduced. The action of T3 at the growth plate may be partially mediated through the FGFR3 pathway. CONCLUSIONS The results suggest that FGFR3 inhibits chondrocyte proliferation by down-regulating TERT expression and reducing telomerase activity indicating an important role for telomerase in sustaining chondrocyte proliferative capacity during bone elongation.
Collapse
Affiliation(s)
- Logan B Smith
- Department of Animal Science, University of California, Davis, CA, 95616, USA.
| | | | | |
Collapse
|
37
|
Ferro F, Spelat R, Beltrami AP, Cesselli D, Curcio F. Isolation and characterization of human dental pulp derived stem cells by using media containing low human serum percentage as clinical grade substitutes for bovine serum. PLoS One 2012; 7:e48945. [PMID: 23155430 PMCID: PMC3498354 DOI: 10.1371/journal.pone.0048945] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 10/02/2012] [Indexed: 01/01/2023] Open
Abstract
Adult stem cells have been proposed as an alternative to embryonic stem cells to study multilineage differentiation in vitro and to use in therapy. Current culture media for isolation and expansion of adult stem cells require the use of large amounts of animal sera, but animal-derived culture reagents give rise to some questions due to the real possibility of infections and severe immune reactions. For these reasons a clinical grade substitute to animal sera is needed. We tested the isolation, proliferation, morphology, stemness related marker expression, and osteoblastic differentiation potential of Dental Pulp Stem Cells (DPSC) in a chemically defined medium containing a low percentage of human serum, 1.25%, in comparison to a medium containing 10% Fetal Bovine Serum (FBS). DPSCs cultured in presence of our isolation/proliferation medium added with low HS percentage were obtained without immune-selection methods and showed high uniformity in the expression of stem cell markers, proliferated at higher rate, and demonstrated comparable osteoblastic potential with respect to DPSCs cultured in 10% FBS. In this study we demonstrated that a chemically defined medium added with low HS percentage, derived from autologous and heterologous sources, could be a valid substitute to FBS-containing media and should be helpful for adult stem cells clinical application.
Collapse
Affiliation(s)
- Federico Ferro
- Department of Medical and Biological Sciences, University of Udine, Udine, Italy.
| | | | | | | | | |
Collapse
|
38
|
Zeng B, Hu J, Yuan R, Hu L, Zhong L, Kang K. Increased expression of importin13 in endometriosis and endometrial carcinoma. Med Sci Monit 2012; 18:CR361-7. [PMID: 22648251 PMCID: PMC3560734 DOI: 10.12659/msm.882879] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background Importin13 (IPO13) is a novel potential marker of corneal epithelial progenitor cells. We investigated the expression and localization of IPO13 in endometrial, endometriotic and endometrial carcinoma tissue. Material/Methods IPO13 expression in endometrial, endometriotic and endometrial carcinoma tissue was examined by immunohistochemistry, qPCR and Western blot. Results Immunohistochemistry studies showed that IPO13 protein was expressed mainly in cytoplasm of glandular epithelial cell and stromal cells. The rate of importin13-positive cells in proliferative phase endometrium was higher (by about 6-fold) than that in secretory endometrium (P<0.05) and the rate of importin13-positive cells in endometriosis and endometrial carcinoma was higher than that in normal secretory phase endometrial tissues (by about 4- and 9-fold, respectively). Immunofluorescence microscopy revealed co-localization of IPO13 with CD34, CD45, c-kit, telomerase, CD90 and CD146. QPCR revealed significantly increased IPO13 mRNA in endometriosis and endometrial carcinoma versus secretory phase endometrium (by about 2- and 10-fold, respectively). Western blot analysis showed that IPO13 protein is enhanced in endometriosis and endometrial carcinoma versus secretory phase endometrium (p<0.05). Conclusions These results demonstrate an increased expression of IPO13 in endometriosis and endometrial carcinoma, which could be involved in the pathogenesis of endometriosis and endometrial carcinoma; IPO13 can serve as an endometrial progenitor/stem cell marker.
Collapse
Affiliation(s)
- Biao Zeng
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Ninagawa N, Murakami R, Isobe E, Tanaka Y, Nakagawa H, Torihashi S. Mesenchymal stem cells originating from ES cells show high telomerase activity and therapeutic benefits. Differentiation 2011; 82:153-64. [PMID: 21855203 DOI: 10.1016/j.diff.2011.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 06/19/2011] [Accepted: 07/13/2011] [Indexed: 02/08/2023]
Abstract
We establish a novel method for the induction and collection of mesenchymal stem cells using a typical cell surface marker, CD105, through adipogenesis from mouse ES cells. ES cells were cultured in a medium for adipogenesis. Mesenchymal stem cells from mouse ES cells were easily identified by the expression of CD105, and were isolated and differentiated into multiple mesenchymal cell types. Mesenchymal stem cells showed remarkable telomerase activity and sustained their growth for a long time with a high potential for differentiation involving skeletal myogenesis in vitro. When mesenchymal stem cells were transplanted into the injured tibialis anterior muscles, they differentiated into skeletal muscle cells in vivo. In addition, they improved the vascular formation, but never formed teratoma for longer than 6 months. Gene expression profiles revealed that mesenchymal stem cells lost pluripotency, while they acquired high potential to differentiate into mesenchymal cell lines. They thus indicate a promising new source of cell-based therapy without teratoma formation.
Collapse
Affiliation(s)
- Nana Ninagawa
- Department of Health Sciences, Nagoya University Graduate School of Medicine, Higashi-ku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
41
|
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: 1.9] [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
|
42
|
Poloni A, Maurizi G, Babini L, Serrani F, Berardinelli E, Mancini S, Costantini B, Discepoli G, Leoni P. Human Mesenchymal Stem Cells from Chorionic Villi and Amniotic Fluid are not Susceptible to Transformation after Extensive in Vitro Expansion. Cell Transplant 2011; 20:643-54. [DOI: 10.3727/096368910x536518] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for cell therapy and tissue engineering. Increasing evidence suggests that MSCs isolated from fetal tissues are more plastic and grow faster than adult MSCs. In this study, we characterized human mesenchymal progenitor cells from chorionic villi (CV) and amniotic fluid (AF) isolated during the first and second trimesters, respectively, and compared them with adult bone marrow-derived MSCs (BM). We evaluated 10 CV, 10 AF, and 6 BM samples expanded until the MSCs reached senescence. We used discarded cells from prenatal analyses for all the experiments. To evaluate the replicative stability of these cells, we studied the telomerase activity, hTERT gene transcription, and telomere length in these cells. Spontaneous chromosomal alterations were excluded by cytogenetic analysis. We studied the expression of c-myc and p53, tumor-associated genes, at different passage in culture and the capacity of these cells to grow in an anchorage-independent manner by using soft agar assay. We isolated homogeneous populations of spindle-shaped CV, AF, and BM cells expressing mesenchymal immunophenotypic markers throughout the period of expansion. CV cells achieved 14 ± 0.9 logs of expansion in 118 days and AF cells achieved 21 ± 0.9 logs in 118 days, while BM cells achieved 11 × 0.4 logs in 84 days. Despite their high proliferation capacity, fetal MSCs showed no telomerase activity, no hTERT and c-myc transcriptions, and maintained long, stable telomeres. A constant expression level of p53 and a normal karyotype were preserved throughout long-term expansion, suggesting the safety of fetal MSCs. In conclusion, our results indicate that fetal MSCs could be an alternative, more accessible resource for cell therapy and regenerative medicine.
Collapse
Affiliation(s)
- Antonella Poloni
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Maurizi
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Lucia Babini
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Federica Serrani
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Eleonora Berardinelli
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Stefania Mancini
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Benedetta Costantini
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| | - Giancarlo Discepoli
- Laboratorio di Citogenetica e Genetica Molecolare, Clinica di Pediatria, Ospedali Riuniti, Ancona, Italy
| | - Pietro Leoni
- Clinica di Ematologia, Dipartimento Scienze Mediche e Chirurgiche, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
43
|
Karaöz E, Okçu A, Gacar G, Sağlam O, Yürüker S, Kenar H. A comprehensive characterization study of human bone marrow mscs with an emphasis on molecular and ultrastructural properties. J Cell Physiol 2011; 226:1367-82. [PMID: 20945392 DOI: 10.1002/jcp.22468] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) continue to draw attention of researchers in the fields of basic science and medicine due to their indispensible regenerative, reparative, angiogenic, anti-apoptotic, and immunosuppressive properties, all of which collectively point out their enormous therapeutic potential. There is still, however, a need for further investigation of their characteristics to broaden their field of use and learn much more about how to control their fate and improve their therapeutic effectiveness. hBM-MSCs were extensively characterized in terms of their growth characteristics, genetic stability, and differentiation capability to the mesodermal and ectodermal cell lineages; a special emphasis was given to their phenotypic and ultrastructural properties. Expression of embryonic stem cell markers Oct4, Rex-1, FoxD-3, Sox2, and Nanog was shown with real-time PCR. Transmission electron microscopy revealed the ultrastructural characteristics of hBM-MSCs; they had pale, irregularly shaped and large euchromatic nuclei, and two distinct areas in their cytoplasm: an intensely stained inner zone rich in mitochondria and rough endoplasmic reticulum (rER) with dilated cisternae and a relatively peripheral zone poor in organelles. hBM-MSCs expressed adipogenic (adipophilin and PPARγ), myogenic (desmin, myogenin, α-SMA), neurogenic (γ-enolase, MAP2a,b, c-fos, nestin, NF-H, NF-L, GFAP, β3-tubulin), osteogenic (osteonectin, osteocalcin, osteopontin, Runx-2, type I collagen), and chondrogenic (type II collagen, SOX9) markers either at RNA or protein level even under basal conditions, without any stimulation towards differentiation. The differentiation potential of hBM-MSCs to adipogenic, osteogenic, and neurogenic lineages was shown by using the relevant differentiation factors.
Collapse
Affiliation(s)
- Erdal Karaöz
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey.
| | | | | | | | | | | |
Collapse
|
44
|
Giardini M, Fernández M, Lira C, Cano M. Leishmania amazonensis: Partial purification and study of the biochemical properties of the telomerase reverse transcriptase activity from promastigote-stage. Exp Parasitol 2011; 127:243-8. [DOI: 10.1016/j.exppara.2010.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 07/28/2010] [Accepted: 08/02/2010] [Indexed: 11/25/2022]
|
45
|
Charbord P. Bone marrow mesenchymal stem cells: historical overview and concepts. Hum Gene Ther 2010; 21:1045-56. [PMID: 20565251 DOI: 10.1089/hum.2010.115] [Citation(s) in RCA: 292] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This review describes the historical emergence of the concept of bone marrow mesenchymal stem cells (MSCs), summarizing data on Wolf and Trentin's hematopoietic inductive microenvironment; Dexter's hematopoiesis-supportive stromal cells; Friedenstein's osteogenic cells; and Pittenger's trilineal osteoblastic, chondrocytic, and adipocytic precursors; to finally introduce the specific bone marrow mesenchymal stem cells with differentiation potential to four lineages (mesenchymal and vascular smooth muscle lineages), and stromal and immunomodulatory capacities. Two points are the object of detailed discussion. The first point envisions the stem cell attributes (multipotentiality, self-renewal, tissue regeneration, population heterogeneity, plasticity, and lineage priming) compared with that of the paradigmatic hematopoietic stem cell. In the second point, we discuss the possible existence of bone marrow cells with greater differentiation potential, eventually pluripotential cells. The latter point raises the issues of cell fusion, reprogramming, or selection under nonstandardized conditions of rare populations of neuroectodermal origin, or of cells that had undergone mesenchymal-to-epithelial transition. In the last section, we review data on MSC senescence and possible malignant transformation secondary to extensive culture, gene transfer of telomerase, or mutations such as leading to Ewing's sarcoma. The set of data leads to the conclusion that bone marrow MSCs constitute a specific adult tissue stem cell population. The multiple characteristics of this stem cell type account for the versatility of the mechanisms of injured tissue repair. Although MSC administration may be extremely useful in a number of clinical applications, their transplantation is not without risks that must not be overlooked when developing cell therapy protocols.
Collapse
Affiliation(s)
- Pierre Charbord
- Institut National de la Recherche et Santé Médicale U, Université Paris XI, Kremlin Bicêtre, France.
| |
Collapse
|
46
|
Dwyer RM, Kerin MJ. Mesenchymal stem cells and cancer: tumor-specific delivery vehicles or therapeutic targets? Hum Gene Ther 2010; 21:1506-12. [PMID: 20649487 DOI: 10.1089/hum.2010.135] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a subset of nonhematopoietic multipotent cells found primarily within the bone marrow stroma. The ability of MSCs to specifically home to sites of tumors and their metastases, while escaping host immune surveillance, holds tremendous promise for tumor-targeted delivery of therapeutic agents. Concerns that MSCs may have an inherent capacity for transformation have led to a number of studies investigating their stability in vitro, as significant ex vivo expansion will be necessary to yield the number of cells required for therapeutic applications. MSCs have also been seen to influence the morphology and proliferation of cells within their vicinity through a combination of cell-to-cell interactions and the secretion of chemoattractant cytokines. Understanding interactions between MSCs and tumor cells is required to support realization of their clinical potential. This review discusses MSCs and cancer in terms of (1) potential for transformation and de novo tumor formation, (2) interactions with epithelial cancer cells in tumor establishment, and (3) potential role after engraftment at the site of an established tumor. Elucidation of any potential negative effect of MSCs in the tumor setting will support development of protocols to minimize these effects while taking full advantage of the remarkable tumor-homing capacity of these cells.
Collapse
Affiliation(s)
- R M Dwyer
- Division of Surgery, School of Medicine, National University of Ireland, Galway, Ireland.
| | | |
Collapse
|
47
|
Tokcaer-Keskin Z, Dikmen ZG, Ayaloglu-Butun F, Gultekin S, Gryaznov SM, Akcali KC. The effect of telomerase template antagonist GRN163L on bone-marrow-derived rat mesenchymal stem cells is reversible and associated with altered expression of cyclin d1, cdk4 and cdk6. Stem Cell Rev Rep 2010; 6:224-33. [PMID: 20180048 DOI: 10.1007/s12015-010-9124-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Telomerase activity is essential for the continued growth and survival of malignant cells, therefore inhibition of this activity presents an attractive target for anti-cancer therapy. The telomerase inhibitor GRN163L, was shown to inhibit the growth of cancer cells both in vitro and in vivo. Mesenchymal stem cells (MSCs) also show telomerase activity in maintaining their self-renewal; therefore the effects of telomerase inhibitors on MSCs may be an issue of concern. MSCs are multipotent cells and are important for the homeostasis of the organism. In this study, we sought to demonstrate in vitro effects of GRN163L on rat MSCs. When MSCs were treated with 1 microM GRN163L, their phenotype changed from spindle-shaped cells to rounded ones and detached from the plate surface, similar to cancer cells. Quantitative-RT-PCR and immunoblotting results revealed that GRN163L holds MSCs at the G1 state of the cell cycle, with a drastic decrease in mRNA and protein levels of cyclin D1 and its cdk counterparts, cdk4 and cdk6. This effect was not observed when MSCs were treated with a mismatch control oligonucleotide. One week after GRN163L was removed, mRNA and protein expressions of the genes, as well as the phenotype of MSCs returned to those of untreated cells. Therefore, we concluded that GRN163L does not interfere with the self-renewal and differentiation of MSCs under short term in vitro culture conditions. Our study provides additional support for treating cancers by administrating GRN163L without depleting the body's stem cell pools.
Collapse
Affiliation(s)
- Zeynep Tokcaer-Keskin
- Department of Molecular Biology and Genetics, Bilkent University, 06800, Bilkent, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
48
|
Singh S, Dhaliwal N, Crawford R, Xiao Y. Cellular senescence and longevity of osteophyte-derived mesenchymal stem cells compared to patient-matched bone marrow stromal cells. J Cell Biochem 2010; 108:839-50. [PMID: 19693768 DOI: 10.1002/jcb.22312] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study aimed to determine the cellular aging of osteophyte-derived mesenchymal cells (oMSCs) in comparison to patient-matched bone marrow stromal cells (bMSCs). Extensive expansion of the cell cultures was performed and early and late passage cells (passages 4 and 9, respectively) were used to study signs of cellular aging, telomere length, telomerase activity, and cell-cycle-related gene expression. Our results showed that cellular aging was more prominent in bMSCs than in oMSCs, and that oMSCs had longer telomere length in late passages compared with bMSCs, although there was no significant difference in telomere lengths in the early passages in either cell type. Telomerase activity was detectable only in early passage oMSCs and not in bMSCs. In osteophyte tissues telomerase-positive cells were found to be located perivascularly and were Stro-1 positive. Fifteen cell-cycle regulator genes were investigated and only three genes (APC, CCND2, and BMP2) were differentially expressed between bMSC and oMSC. Our results indicate that oMSCs retain a level of telomerase activity in vitro, which may account for the relatively greater longevity of these cells, compared with bMSCs, by preventing replicative senescence.
Collapse
Affiliation(s)
- Sanjleena Singh
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | | | | | | |
Collapse
|
49
|
Karaöz E, Doğan BN, Aksoy A, Gacar G, Akyüz S, Ayhan S, Genç ZS, Yürüker S, Duruksu G, Demircan PÇ, Sarıboyacı AE. Isolation and in vitro characterisation of dental pulp stem cells from natal teeth. Histochem Cell Biol 2009; 133:95-112. [PMID: 19816704 DOI: 10.1007/s00418-009-0646-5] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2009] [Indexed: 01/06/2023]
|
50
|
Recent insights into the molecular mechanisms involved in aging and the malignant transformation of adult stem/progenitor cells and their therapeutic implications. Ageing Res Rev 2009; 8:94-112. [PMID: 19114129 DOI: 10.1016/j.arr.2008.12.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 12/04/2008] [Indexed: 02/07/2023]
Abstract
Recent advancements in tissue-resident adult stem/progenitor cell research have revealed that enhanced telomere attrition, oxidative stress, ultraviolet radiation exposure and oncogenic events leading to severe DNA damages and genomic instability may occur in these immature and regenerative cells during chronological aging. Particularly, the alterations in key signaling components controlling their self-renewal capacity and an up-regulation of tumor suppressor gene products such as p16(INK4A), p19(ARF), ataxia-telangiectasia mutated (ATM) kinase, p53 and/or the forkhead box O (FOXOs) family of transcription factors may result in their dysfunctions, growth arrest and senescence or apoptotic death during the aging process. These molecular events may culminate in a progressive decline in the regenerative functions and the number of tissue-resident adult stem/progenitor cells, and age-related disease development. Conversely, the telomerase re-activation and accumulation of numerous genetic and/or epigenetic alterations in adult stem/progenitor cells with advancing age may result in their immortalization and malignant transformation into highly leukemic or tumorigenic cancer-initiating cells and cancer initiation. Therefore, the cell-replacement and gene therapies and molecular targeting of aged and dysfunctional adult stem/progenitor cells including their malignant counterpart, cancer-initiating cells, hold great promise for treating and even curing diverse devastating human diseases. These diseases include premature aging diseases, hematopoietic, cardiovascular, musculoskeletal, pulmonary, ocular, urogenital, neurodegenerative and skin disorders and aggressive and recurrent cancers.
Collapse
|