1
|
Mendiratta M, Mohanty S, Sahoo RK. Umbilical Cord-Derived Mesenchymal Stromal Cells For Prevention of Chronic Graft-vs-Host Disease. JAMA Oncol 2024; 10:986-987. [PMID: 38814632 DOI: 10.1001/jamaoncol.2024.1521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Affiliation(s)
- Mohini Mendiratta
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
2
|
Daks A, Parfenyev S, Shuvalov O, Fedorova O, Nazarov A, Melino G, Barlev NA. Lysine-specific methyltransferase Set7/9 in stemness, differentiation, and development. Biol Direct 2024; 19:41. [PMID: 38812048 PMCID: PMC11137904 DOI: 10.1186/s13062-024-00484-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024] Open
Abstract
The enzymes performing protein post-translational modifications (PTMs) form a critical post-translational regulatory circuitry that orchestrates literally all cellular processes in the organism. In particular, the balance between cellular stemness and differentiation is crucial for the development of multicellular organisms. Importantly, the fine-tuning of this balance on the genetic level is largely mediated by specific PTMs of histones including lysine methylation. Lysine methylation is carried out by special enzymes (lysine methyltransferases) that transfer the methyl group from S-adenosyl-L-methionine to the lysine residues of protein substrates. Set7/9 is one of the exemplary protein methyltransferases that however, has not been fully studied yet. It was originally discovered as histone H3 lysine 4-specific methyltransferase, which later was shown to methylate a number of non-histone proteins that are crucial regulators of stemness and differentiation, including p53, pRb, YAP, DNMT1, SOX2, FOXO3, and others. In this review we summarize the information available to date on the role of Set7/9 in cellular differentiation and tissue development during embryogenesis and in adult organisms. Finally, we highlight and discuss the role of Set7/9 in pathological processes associated with aberrant cellular differentiation and self-renewal, including the formation of cancer stem cells.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064.
| | - Sergey Parfenyev
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Alexander Nazarov
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, St Petersburg, Russian Federation, 194064.
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, 001000, Astana, Kazakhstan.
| |
Collapse
|
3
|
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
|
4
|
Panda D, Nayak S. Stem Cell-Based Tissue Engineering Approaches for Diabetic Foot Ulcer: a Review from Mechanism to Clinical Trial. Stem Cell Rev Rep 2024; 20:88-123. [PMID: 37867186 DOI: 10.1007/s12015-023-10640-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 10/24/2023]
Abstract
Diabetic foot ulcer (DFU) is a complication from incomplete or prolonged wound healing, at times requires amputation, putting substantial health and socioeconomic burden. Wound healing is a dynamic overlapping process that can be regulated by arrays of molecular factors showing redundancy in function. However, dysregulation in the mechanism of angiogenesis, extra cellular matrix (ECM) formation and immune modulation are the major causes for impair wound healing in hyperglycaemic patients. Despite development of wound care research, there is a lack of well-accepted targeted therapy with multidisciplinary approach for DFU treatment. Stem cell therapy holds a promising outcome both in preclinical and clinical trials because of its ability to promote healing via regeneration and specialized tissue differentiation. Among different types of stem cells, regenerative potential of mesenchymal stem cell (MSC) is well demonstrated in both experimental and clinical trial. Still there is a huge knowledge gap among medical practitioners for deciding the best stem cell source, administration route, and safety. This review strengthens the fact that why stem cell therapy is a promising candidate to treat DFU and cited multiple tissue engineering and biomaterial-based approaches for delivering stem cells and their aftermath paracrine events. Based on the pre-clinical and clinical studies, the review tried to come up with optimum stem cell source and delivery route for the treatment of DFU. At last, the review glances on possible direction to enhance therapeutics strategy for the same, including different approaches like: phytocompounds, exosomes, scaffold geometry, cell preconditioning and licensing etc.
Collapse
Affiliation(s)
- Debarchan Panda
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Sunita Nayak
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
5
|
Manousakis E, Miralles CM, Esquerda MG, Wright RHG. CDKN1A/p21 in Breast Cancer: Part of the Problem, or Part of the Solution? Int J Mol Sci 2023; 24:17488. [PMID: 38139316 PMCID: PMC10743848 DOI: 10.3390/ijms242417488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Cyclin-dependent kinase inhibitor 1A (Cip1/Waf1/CDKN1A/p21) is a well-established protein, primarily recognised for its pivotal role in the cell cycle, where it induces cell cycle arrest by inhibiting the activity of cyclin-dependent kinases (CDKs). Over the years, extensive research has shed light on various additional mechanisms involving CDKN1A/p21, implicating it in processes such as apoptosis, DNA damage response (DDR), and the regulation of stem cell fate. Interestingly, p21 can function either as an oncogene or as a tumour suppressor in these contexts. Complicating matters further, the expression of CDKN1A/p21 is elevated in certain tumour types while downregulated in others. In this comprehensive review, we provide an overview of the multifaceted functions of CDKN1A/p21, present clinical data pertaining to cancer patients, and delve into potential strategies for targeting CDKN1A/p21 as a therapeutic approach to cancer. Manipulating CDKN1A/p21 shows great promise for therapy given its involvement in multiple cancer hallmarks, such as sustained cell proliferation, the renewal of cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cell migration, and resistance to chemotherapy. Given the dual role of CDKN1A/p21 in these processes, a more in-depth understanding of its specific mechanisms of action and its regulatory network is imperative to establishing successful therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Roni H. G. Wright
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Barcelona, Spain
| |
Collapse
|
6
|
Lenz LS, Wink MR. The other side of the coin: mesenchymal stromal cell immortalization beyond evasion of senescence. Hum Cell 2023; 36:1593-1603. [PMID: 37341871 DOI: 10.1007/s13577-023-00925-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/23/2023] [Indexed: 06/22/2023]
Abstract
Mesenchymal stromal cells (MSC) are promising options to cellular therapy to several clinical disorders, mainly because of its ability to immunomodulate and differentiate into different cell types. Even though MSC can be isolated from different sources, a major challenge to understanding the biological effects is that the primary cells undergo replicative senescence after a limited number of cell divisions in culture, requiring time-consuming and technically challenging approaches to get a sufficient cell number for clinical applications. Therefore, a new isolation, characterization, and expansion is necessary every time, which increases the variability and is time-consuming. Immortalization is a strategy that can overcome these challenges. Therefore, here, we review the different methodologies available to cellular immortalization, and discuss the literature regarding MSC immortalization and the broader biological consequences that extend beyond the mere increase in proliferation potential.
Collapse
Affiliation(s)
- Luana Suéling Lenz
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil
| | - Márcia Rosângela Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil.
- Departamento de Ciências Básicas da Saúde (DCBS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
7
|
Yang X, Wang Y, Rovella V, Candi E, Jia W, Bernassola F, Bove P, Piacentini M, Scimeca M, Sica G, Tisone G, Mauriello A, Wei L, Melino G, Shi Y. Aged mesenchymal stem cells and inflammation: from pathology to potential therapeutic strategies. Biol Direct 2023; 18:40. [PMID: 37464416 PMCID: PMC10353240 DOI: 10.1186/s13062-023-00394-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
Natural ageing of organisms and corresponding age-related diseases result mainly from stem cell ageing and "inflammaging". Mesenchymal stem cells (MSCs) exhibit very high immune-regulating capacity and are promising candidates for immune-related disease treatment. However, the effect of MSC application is not satisfactory for some patients, especially in elderly individuals. With ageing, MSCs undergo many changes, including altered cell population reduction and differentiation ability, reduced migratory and homing capacity and, most important, defective immunosuppression. It is necessary to explore the relationship between the "inflammaging" and aged MSCs to prevent age-related diseases and increase the therapeutic effects of MSCs. In this review, we discuss changes in naturally ageing MSCs mainly from an inflammation perspective and propose some ideas for rejuvenating aged MSCs in future treatments.
Collapse
Affiliation(s)
- Xue Yang
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu China
| | - Ying Wang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu China
| | - Valentina Rovella
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Wei Jia
- Center for Translational Medicine, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233 China
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong China
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Pierluigi Bove
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Mauro Piacentini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Giuseppe Sica
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Giuseppe Tisone
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Lixin Wei
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438 China
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, 00133 Italy
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu China
| |
Collapse
|
8
|
Romecín PA, Vinyoles M, López-Millán B, de la Guardia RD, Atucha NM, Querol S, Bueno C, Benitez R, Gonzalez-Rey E, Delgado M, Menéndez P. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:88-96. [PMID: 35641173 PMCID: PMC8895490 DOI: 10.1093/stcltm/szab007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stromal stem/cells (MSC) therapies are clinically used in a wide range of disorders based on their robust HLA-independent immunosuppressive and anti-inflammatory properties. However, the mechanisms underlying MSC therapeutic activity remain elusive as demonstrated by the unpredictable therapeutic efficacy of MSC infusions reported in multiple clinical trials. A seminal recent study showed that infused MSCs are actively induced to undergo apoptosis by recipient cytotoxic T cells, a mechanism that triggers in vivo recipient-induced immunomodulation by such apoptotic MSCs, and the need for such recipient cytotoxic cell activity could be replaced by the administration of ex vivo-generated apoptotic MSCs. Moreover, the use of MSC-derived extracellular vesicles (MSC-EVs) is being actively explored as a cell-free therapeutic alternative over the parental MSCs. We hypothesized that the introduction of a “suicide gene” switch into MSCs may offer on-demand in vivo apoptosis of transplanted MSCs. Here, we prompted to investigate the utility of the iCasp9/AP1903 suicide gene system in inducing apoptosis of MSCs. iCasp9/AP1903-induced apoptotic MSCs (MSCiCasp9+) were tested in vitro and in in vivo models of acute colitis. Our data show a very similar and robust immunosuppressive and anti-inflammatory properties of both “parental” alive MSCGFP+ cells and apoptotic MSCiCasp9+ cells in vitro and in vivo regardless of whether apoptosis was induced in vivo or in vitro before administering MSCiCasp9+ lysates. This development of an efficient iCasp9 switch may potentiate the safety of MSC-based therapies in the case of an adverse event and, will also circumvent current logistic technical limitations and biological uncertainties associated to MSC-EVs.
Collapse
Affiliation(s)
- Paola Alejandra Romecín
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- Paola Alejandra Romecin, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Carrer Casanova 143, 4º floor, 08036, Barcelona, Spain. Tel: (+34) 93 5572810;
| | | | - Belén López-Millán
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- GENYO, Centro Pfizer-Universidad de Granada-Junta de Andalucía de Genómica e Investigación Oncológica, Granada, Spain
| | - Rafael Diaz de la Guardia
- GENYO, Centro Pfizer-Universidad de Granada-Junta de Andalucía de Genómica e Investigación Oncológica, Granada, Spain
| | - Noemi M Atucha
- Departamento de Fisiologia Humana, Facultad de Medicina, Murcia, Spain
| | - Sergi Querol
- RICORS-TERAV, ISCIII, Madrid, Spain
- Banc de Sang i Teixits, Barcelona, Spain
| | - Clara Bueno
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- CIBERONC, ISCIII, Barcelona, Spain
| | - Raquel Benitez
- Instituto de Parasitologia y Biomedicina López-Neyra (IPBLN-CSIC), Armilla, Granada, Spain
| | - Elena Gonzalez-Rey
- Instituto de Parasitologia y Biomedicina López-Neyra (IPBLN-CSIC), Armilla, Granada, Spain
| | - Mario Delgado
- Instituto de Parasitologia y Biomedicina López-Neyra (IPBLN-CSIC), Armilla, Granada, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- CIBERONC, ISCIII, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Corresponding author: Pablo Menéndez, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Carrer Casanova 143, 4º floor, 08036, Barcelona, Spain. Tel: (+34) 93 5572810;
| |
Collapse
|
9
|
Marinescu CI, Preda MB, Neculachi CA, Rusu EG, Popescu S, Burlacu A. Identification of a Hematopoietic Cell Population Emerging From Mouse Bone Marrow With Proliferative Potential In Vitro and Immunomodulatory Capacity. Front Immunol 2021; 12:698070. [PMID: 34413852 PMCID: PMC8368722 DOI: 10.3389/fimmu.2021.698070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/16/2021] [Indexed: 11/13/2022] Open
Abstract
There is continuing interest in therapeutic applications of bone marrow-derived mesenchymal stromal cells (MSC). Unlike human counterparts, mouse MSC are difficult to propagate in vitro due to their contamination with adherent hematopoietic cells that overgrow the cultures. Here we investigated the properties of these contaminating cells, referred to as bone marrow-derived proliferating hematopoietic cells (BM-PHC). The results showed that both BM-PHC and MSC had strong immunomodulatory properties on T cells in vitro, with PGE2 and NO involved in this mechanism. However, BM-PHC were stronger immunomodulators than MSC, with CCL-6 identified as putative molecule responsible for superior effects. In vivo studies showed that, in contrast to BM-PHC, MSC endorsed a more rapid xenograft tumor rejection, thus indicating a particular context in which only MSC therapy would produce positive outcomes. In conclusion, bone marrow contains two cell populations with immunomodulatory properties, which are valuable sources for therapeutic studies in specific disease-relevant contexts.
Collapse
Affiliation(s)
- Catalina-Iolanda Marinescu
- Laboratory of Stem Cell Biology, Department of Regenerative Medicine, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Mihai Bogdan Preda
- Laboratory of Stem Cell Biology, Department of Regenerative Medicine, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Carmen Alexandra Neculachi
- Laboratory of Stem Cell Biology, Department of Regenerative Medicine, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Evelyn Gabriela Rusu
- Laboratory of Stem Cell Biology, Department of Regenerative Medicine, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Sinziana Popescu
- Laboratory of Stem Cell Biology, Department of Regenerative Medicine, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Alexandrina Burlacu
- Laboratory of Stem Cell Biology, Department of Regenerative Medicine, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
10
|
Yadahalli R, Kheur S, Adwani A, Bhonde R, Raj AT, Patil S. Nuclear Blebbing Frequency in Tobacco-Induced Oral Potentially Malignant Disorders: A Pilot Study. Acta Cytol 2021; 65:403-410. [PMID: 34120116 DOI: 10.1159/000516496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/10/2021] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Tobacco contains several genotoxic agents including N-nitrosamine which has the potential to cause significant nuclear damage. Nuclear blebbing is a form of protrusion on the nuclear membrane and could potentially be caused by tobacco-induced genotoxicity and is closely associated with malignancy. Thus, the present study aimed to assess if tobacco-associated oral potentially malignant disorders including oral submucous fibrosis (OSF) and oral leukoplakia have a higher nuclear blebbing frequency than patients with normal oral mucosa with no history of tobacco use. METHODS The sample consisted of patients with OSF (n = 30) and oral leukoplakia (n = 10) and normal oral mucosa (n = 10). Exfoliated cells collected from the study groups were smeared on a clean microscopic slide and stained by May-Grunwald-Giemsa stain. A baseline frequency of nuclear blebbing was evaluated using a bright-field microscope with a ×100 objective. The number of nuclear blebbing per 1,000 epithelial cells was recorded and expressed in percentage. ANOVA, the Mann-Whitney U test, and Spearman's correlation were used to analyze the data. RESULTS The mean rank of distribution of nuclear blebbing showed significant difference between all 3 groups, with the highest frequency noted in leukoplakia, followed by oral submucous and normal oral mucosa. Within OSF, the frequency of nuclear blebbing significantly increased from early stage to advanced stage. In OSF, a statistically significant positive linear correlation was noted between duration (in years), frequency (per day) of tobacco use, clinical grading, and nuclear blebbing. DISCUSSION/CONCLUSIONS The frequency of nuclear blebbing was significantly higher in oral potentially malignant disorders than normal mucosa. Nuclear blebbing also exhibited a strong dose- and time-dependent correlation with tobacco usage and clinical staging in OSF. The nuclear blebbing frequency could be a noninvasive, economic tool to assess malignant risk in tobacco-induced oral potentially malignant disorders.
Collapse
Affiliation(s)
- Roopa Yadahalli
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Supriya Kheur
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Aanchal Adwani
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - Ramesh Bhonde
- Department of Oral Pathology and Microbiology, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, India
| | - A Thirumal Raj
- Department of Oral Pathology and Microbiology, Sri Venkateswara Dental College and Hospital, Chennai, India
| | - Shankargouda Patil
- Division of Oral Pathology College of Dentistry, Department of Maxillofacial Surgery and Diagnostic Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
11
|
Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol 2021; 14:24. [PMID: 33579329 PMCID: PMC7880217 DOI: 10.1186/s13045-021-01037-x] [Citation(s) in RCA: 299] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs), also known as mesenchymal stem cells, have been intensely investigated for clinical applications within the last decades. However, the majority of registered clinical trials applying MSC therapy for diverse human diseases have fallen short of expectations, despite the encouraging pre-clinical outcomes in varied animal disease models. This can be attributable to inconsistent criteria for MSCs identity across studies and their inherited heterogeneity. Nowadays, with the emergence of advanced biological techniques and substantial improvements in bio-engineered materials, strategies have been developed to overcome clinical challenges in MSC application. Here in this review, we will discuss the major challenges of MSC therapies in clinical application, the factors impacting the diversity of MSCs, the potential approaches that modify MSC products with the highest therapeutic potential, and finally the usage of MSCs for COVID-19 pandemic disease.
Collapse
|
12
|
Ghatak D, Das Ghosh D, Roychoudhury S. Cancer Stemness: p53 at the Wheel. Front Oncol 2021; 10:604124. [PMID: 33505918 PMCID: PMC7830093 DOI: 10.3389/fonc.2020.604124] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor suppressor p53 maintains an equilibrium between self-renewal and differentiation to sustain a limited repertoire of stem cells for proper development and maintenance of tissue homeostasis. Inactivation of p53 disrupts this balance and promotes pluripotency and somatic cell reprogramming. A few reports in recent years have indicated that prevalent TP53 oncogenic gain-of-function (GOF) mutations further boosts the stemness properties of cancer cells. In this review, we discuss the role of wild type p53 in regulating pluripotency of normal stem cells and various mechanisms that control the balance between self-renewal and differentiation in embryonic and adult stem cells. We also highlight how inactivating and GOF mutations in p53 stimulate stemness in cancer cells. Further, we have explored the various mechanisms of mutant p53-driven cancer stemness, particularly emphasizing on the non-coding RNA mediated epigenetic regulation. We have also analyzed the association of cancer stemness with other crucial gain-of-function properties of mutant p53 such as epithelial to mesenchymal transition phenotypes and chemoresistance to understand how activation of one affects the other. Given the critical role of cancer stem-like cells in tumor maintenance, cancer progression, and therapy resistance of mutant p53 tumors, targeting them might improve therapeutic efficacy in human cancers with TP53 mutations.
Collapse
Affiliation(s)
- Dishari Ghatak
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Damayanti Das Ghosh
- Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| | - Susanta Roychoudhury
- Division of Research, Saroj Gupta Cancer Centre and Research Institute, Kolkata, India
| |
Collapse
|
13
|
Budgude P, Kale V, Vaidya A. Mesenchymal stromal cell‐derived extracellular vesicles as cell‐free biologics for the ex vivo expansion of hematopoietic stem cells. Cell Biol Int 2020; 44:1078-1102. [DOI: 10.1002/cbin.11313] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pallavi Budgude
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell ResearchSymbiosis International (Deemed University) Pune 412115 India
- Symbiosis School of Biological SciencesSymbiosis International (Deemed University) Pune 412115 India
| |
Collapse
|
14
|
Yao L, Yu F, Xu Y, Wang Y, Zuo Y, Wang C, Ye L. DNA damage response manages cell cycle restriction of senile multipotent mesenchymal stromal cells. Mol Biol Rep 2019; 47:809-818. [PMID: 31664596 DOI: 10.1007/s11033-019-05150-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/18/2019] [Indexed: 02/05/2023]
Abstract
Multipotent mesenchymal stromal cells (MMSCs) are promising to treat a variety of traumatic and degenerative diseases. However, in vitro-passage aging induces cell cycle arrest and a series of genetic and biological changes, which greatly limits ex vivo cell number expansion and further clinical application of MMSCs. In most cases, DNA damage and DNA damage response (DDR) act as the main cause and executor of cellular senescence respectively. Mechanistically, DNA damage signals induce cell cycle arrest and DNA damage repair via DDR. If the DNA damage is indelible, MMSCs would entry into a permanent cell cycle arrest. It should be noted that apart from DDR signaling, certain proliferation or metabolism pathways are also occupied in DNA damage related cell cycle arrest. New findings of these aspects will also be summarized in this study. In summary, we aim to provide a comprehensive review of DDR associated cell cycle regulation and other major molecular signaling in the senescence of MMSCs. Above knowledge could contribute to improve the limited capacity of in vitro expansion of MMSCs, and then promote their clinical applications.
Collapse
Affiliation(s)
- Lin Yao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yining Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yitian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanqin Zuo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. .,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Khan RS, Newsome PN. A Comparison of Phenotypic and Functional Properties of Mesenchymal Stromal Cells and Multipotent Adult Progenitor Cells. Front Immunol 2019; 10:1952. [PMID: 31555259 PMCID: PMC6724467 DOI: 10.3389/fimmu.2019.01952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022] Open
Abstract
Both Multipotent Adult Progenitor Cells and Mesenchymal Stromal Cells are bone-marrow derived, non-haematopoietic adherent cells, that are well-known for having immunomodulatory and pro-angiogenic properties, whilst being relatively non-immunogenic. However, they are phenotypically and functionally distinct cell types, which has implications for their efficacy in different settings. In this review we compare the phenotypic and functional properties of these two cell types, to help in determining which would be the superior cell type for different applications.
Collapse
Affiliation(s)
- Reenam S Khan
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
16
|
Relevance of Non-Targeted Effects for Radiotherapy and Diagnostic Radiology; A Historical and Conceptual Analysis of Key Players. Cancers (Basel) 2019; 11:cancers11091236. [PMID: 31450803 PMCID: PMC6770832 DOI: 10.3390/cancers11091236] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 11/17/2022] Open
Abstract
Non-targeted effects (NTE) such as bystander effects or genomic instability have been known for many years but their significance for radiotherapy or medical diagnostic radiology are far from clear. Central to the issue are reported differences in the response of normal and tumour tissues to signals from directly irradiated cells. This review will discuss possible mechanisms and implications of these different responses and will then discuss possible new therapeutic avenues suggested by the analysis. Finally, the importance of NTE for diagnostic radiology and nuclear medicine which stems from the dominance of NTE in the low-dose region of the dose–response curve will be presented. Areas such as second cancer induction and microenvironment plasticity will be discussed.
Collapse
|
17
|
Abstract
Mesenchymal Stem Cells (MSCs) are a heterogeneous population of fibroblast-like cells which maintain self-renewability and pluripotency to differentiate into mesodermal cell lineages. The use of MSCs in clinical settings began with high enthusiasm and the number of MSC-based clinical trials has been rising ever since. However; the very unique characteristics of MSCs that made them suitable to for therapeutic use, might give rise to unwanted outcomes, including tumor formation and progression. In this paper, we present a model of carcinogenesis initiated by MSCs, which chains together the tissue organization field theory, the stem cell theory, and the inflammation-cancer chain. We believe that some tissue resident stem cells could be leaked cells from bone marrow MSC pool to various injured tissue, which consequently transform and integrate in the host tissue. If the injury persists or chronic inflammation develops, as a consequence of recurring exposure to growth factors, cytokines, etc. the newly formed tissue from MSCs, which still has conserved their mesenchymal and stemness features, go through rapid population expansion, and nullify their tumor suppressor genes, and hence give rise to neoplastic cell (carcinomas, sarcomas, and carcino-sarcomas). Considering the probability of this hypothesis being true, the clinical and therapeutic use of MSCs should be with caution, and the recipients' long term follow-up seems to be insightful.
Collapse
|
18
|
Abarrategi A, Gambera S, Alfranca A, Rodriguez-Milla MA, Perez-Tavarez R, Rouault-Pierre K, Waclawiczek A, Chakravarty P, Mulero F, Trigueros C, Navarro S, Bonnet D, García-Castro J. c-Fos induces chondrogenic tumor formation in immortalized human mesenchymal progenitor cells. Sci Rep 2018; 8:15615. [PMID: 30353072 PMCID: PMC6199246 DOI: 10.1038/s41598-018-33689-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal progenitor cells (MPCs) have been hypothesized as cells of origin for sarcomas, and c-Fos transcription factor has been showed to act as an oncogene in bone tumors. In this study, we show c-Fos is present in most sarcomas with chondral phenotype, while multiple other genes are related to c-Fos expression pattern. To further define the role of c-Fos in sarcomagenesis, we expressed it in primary human MPCs (hMPCs), immortalized hMPCs and transformed murine MPCs (mMPCs). In immortalized hMPCs, c-Fos expression generated morphological changes, reduced mobility capacity and impaired adipogenic- and osteogenic-differentiation potentials. Remarkably, immortalized hMPCs or mMPCs expressing c-Fos generated tumors harboring a chondrogenic phenotype and morphology. Thus, here we show that c-Fos protein has a key role in sarcomas and that c-Fos expression in immortalized MPCs yields cell transformation and chondrogenic tumor formation.
Collapse
Affiliation(s)
- Ander Abarrategi
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, E-28021, Spain
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, WC2A 3LY, UK
| | - Stefano Gambera
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, E-28021, Spain
| | - Arantzazu Alfranca
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, E-28021, Spain
| | | | | | - Kevin Rouault-Pierre
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, WC2A 3LY, UK
| | - Alexander Waclawiczek
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, WC2A 3LY, UK
| | - Probir Chakravarty
- Bioinformatics Core, The Francis Crick Institute, London, United Kingdom
| | - Francisca Mulero
- Molecular Image Core Unit, Spanish National Cancer Research Centre, Madrid, E-28029, Spain
| | - César Trigueros
- Mesenchymal and Hematopoietic Stem Cell Laboratory, Fundación Inbiomed, San Sebastian, E-20009, Spain
| | - Samuel Navarro
- Pathology Department, University of Valencia, Valencia, E-46010, Spain
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, WC2A 3LY, UK
| | - Javier García-Castro
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Madrid, E-28021, Spain.
| |
Collapse
|
19
|
Phermthai T, Pokathikorn P, Wichitwiengrat S, Thongbopit S, Tungprasertpol K, Julavijitphong S. P53 Mutation and Epigenetic Imprinted IGF2/H19 Gene Analysis in Mesenchymal Stem Cells Derived from Amniotic Fluid, Amnion, Endometrium, and Wharton's Jelly. Stem Cells Dev 2017. [PMID: 28629288 DOI: 10.1089/scd.2016.0356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSC) are promising cells for medical therapy. In in vitro expansion, MSC can give rise to progeny with genomic and epigenomic alterations, resulting in senescence, loss of terminal differentiation, and transformation to cancer. However, MSC genome protects its genetic instability by a guardian function of the P53 tumor suppressor gene and epigenetic balance system during MSC culture. Mutations of P53 and epigenetic alterations have been reported to disrupt the quality and quantity of MSC and initiate tumorigenesis. We monitor P53 and epigenetic changes in MSC derived from amniotic fluid (AF-MSC), amnion membrane (AM-MSC), endometrium (EM-MSC), and Wharton's jelly (WJ-MSC) by the missense mutation analysis of the P53 gene and the expression levels of P53, and epigenetic insulin-like growth factor 2 (IGF2) and H19-imprinted genes. Our work demonstrates a variation of P53 expression among different MSC types. AF-MSC has a high P53 expression level with retaining a stability of P53 expression throughout a long culture period, whereas EM-MSC and WJ-MSC showed variation of P53 gene expression during culture. Epigenetic analysis showed a stable H19 expression pattern in AF-MSC, AM-MSC, and EM-MSC culture, whereas H19 expression fluctuated in WJ-MSC culture. We conclude that gene instability can be found during in vitro MSC expansion. With awareness to MSC quality and safety in MSC transformation risk, P53 mutation and IGF2 and H19-imprinted gene analysis should be applied to monitor in therapeutic-grade MSC. We also demonstrated that AF-MSC is one of the most interesting MSC for medical therapy because of its high genomic stability and epigenetic fidelity.
Collapse
Affiliation(s)
- Tatsanee Phermthai
- Stem Cell Research and Development Unit, Obstetrics and Gynecology Department, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Puttachart Pokathikorn
- Stem Cell Research and Development Unit, Obstetrics and Gynecology Department, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Suparat Wichitwiengrat
- Stem Cell Research and Development Unit, Obstetrics and Gynecology Department, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Sasiprapa Thongbopit
- Stem Cell Research and Development Unit, Obstetrics and Gynecology Department, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Kittima Tungprasertpol
- Stem Cell Research and Development Unit, Obstetrics and Gynecology Department, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Suphakde Julavijitphong
- Stem Cell Research and Development Unit, Obstetrics and Gynecology Department, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| |
Collapse
|
20
|
cFOS-SOX9 Axis Reprograms Bone Marrow-Derived Mesenchymal Stem Cells into Chondroblastic Osteosarcoma. Stem Cell Reports 2017; 8:1630-1644. [PMID: 28552607 PMCID: PMC5470112 DOI: 10.1016/j.stemcr.2017.04.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 01/13/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) are proposed as the cells of origin of several subtypes of osteosarcoma (OS). However, signals that direct BMSCs to form different subtypes of OS are unclear. Here we show that the default tumor type from spontaneously transformed p53 knockout (p53_KO) BMSCs is osteoblastic OS. The development of this default tumor type caused by p53 loss can be overridden by various oncogenic signals: RAS reprograms p53_KO BMSCs into undifferentiated sarcoma, AKT enhances osteoblastic OS, while cFOS promotes chondroblastic OS formation. We focus on studying the mechanism of cFOS-induced chondroblastic OS formation. Integrated genome-wide studies reveal a regulatory mechanism whereby cFOS binds to the promoter of a key chondroblastic transcription factor, Sox9, and induces its transcription in BMSCs. Importantly, SOX9 mediates cFOS-induced cartilage formation in chondroblastic OS. In summary, oncogenes determine tumor types derived from BMSCs, and the cFOS-SOX9 axis is critical for chondroblastic OS formation. The default tumor type from p53_KO BMSCs is osteoblastic OS Oncogenes reprogram p53_KO BMSCs into different sarcomas cFOS promotes chondroblastic OS from p53_KO BMSCs SOX9 is a mediator of cFOS in promoting chondroblastic OS
Collapse
|
21
|
Molchadsky A, Rotter V. p53 and its mutants on the slippery road from stemness to carcinogenesis. Carcinogenesis 2017; 38:347-358. [PMID: 28334334 DOI: 10.1093/carcin/bgw092] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/25/2016] [Indexed: 12/18/2022] Open
Abstract
Normal development, tissue homeostasis and regeneration following injury rely on the proper functions of wide repertoire of stem cells (SCs) persisting during embryonic period and throughout the adult life. Therefore, SCs employ robust mechanisms to preserve their genomic integrity and avoid heritage of mutations to their daughter cells. Importantly, propagation of SCs with faulty DNA as well as dedifferentiation of genomically altered somatic cells may result in derivation of cancer SCs, which are considered to be the driving force of the tumorigenic process. Multiple experimental evidence suggest that p53, the central tumor suppressor gene, plays a critical regulatory role in determination of SCs destiny, thereby eliminating damaged SCs from the general SC population. Notably, mutant p53 proteins do not only lose the tumor suppressive function, but rather gain new oncogenic function that markedly promotes various aspects of carcinogenesis. In this review, we elaborate on the role of wild type and mutant p53 proteins in the various SCs types that appear under homeostatic conditions as well as in cancer. It is plausible that the growing understanding of the mechanisms underlying cancer SC phenotype and p53 malfunction will allow future optimization of cancer therapeutics in the context of precision medicine.
Collapse
Affiliation(s)
- Alina Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
22
|
Husak Z, Dworzak MN. Chronic stress induces CD99, suppresses autophagy, and affects spontaneous adipogenesis in human bone marrow stromal cells. Stem Cell Res Ther 2017; 8:83. [PMID: 28420430 PMCID: PMC5395812 DOI: 10.1186/s13287-017-0532-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone marrow-derived mesenchymal stromal cells (MSCs) are multipotent cells with a high constitutive level of autophagy and low expression of CD99. Under certain conditions, MSCs may develop tumorigenic properties. However, these transformation-induced conditions are largely unknown. Recently, we have identified an association between Hsp70, a main participant in cellular stress response and tumorigenesis, and CD99. Preliminary observations had revealed upregulation of both proteins in stressed long-term cultured MSCs. And so we hypothesized that CD99 is implicated in stress-induced mechanisms of cellular transformation in MSCs. Hence, we investigated the effects of prolonged stress on MSCs and the role of CD99 and autophagy in their survival. METHODS Human telomerase reverse transcriptase (hTERT) overexpressing immortalized MSCs and primary bone marrow stromal cells were used to investigate the influence of long-term serum deprivation and hypoxia on growth and differentiation of MSCs. Cell proliferation and apoptosis were evaluated using flow cytometry, differentiation capabilities of MSCs were assessed by immunohistochemical staining followed by microscopic examination. CD99, Hsp70 expression were analyzed using flow cytometry, western blotting, and reverse transcriptase polymerase chain reaction. Autophagy was explored with specific inhibitors using cell morphology examination and western blotting. RESULTS Chronic stress factors are able to change the morphology of MSCs and to inhibit spontaneous differentiation into adipocyte lineage. Furthermore, CD99 elevation and downregulation of p53 and p21 accompanied defective autophagy, which is usually associated with tumor formation. We found that inhibition of autophagy by chloroquine promoted cell detachment and modulated CD99 expression level whereas incorporation of CD99 recombinant protein into the cells suppressed autophagy. CONCLUSIONS Obtained results provide a model for chronic stress-induced transformation of MSCs via CD99 and may therefore be highly relevant to mesenchymal tumorigenesis.
Collapse
Affiliation(s)
- Zvenyslava Husak
- St. Anna Kinderkrebsforschung, Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Michael N. Dworzak
- St. Anna Kinderkrebsforschung, Children’s Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
- St. Anna Kinderspital, Kinderspitalgasse 6, 1090 Vienna, Austria
| |
Collapse
|
23
|
Lv B, Li F, Fang J, Xu L, Sun C, Han J, Hua T, Zhang Z, Feng Z, Jiang X. Hypoxia inducible factor 1α promotes survival of mesenchymal stem cells under hypoxia. Am J Transl Res 2017; 9:1521-1529. [PMID: 28386377 PMCID: PMC5376042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/18/2016] [Indexed: 06/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are ideal materials for cell therapy. Research has indicated that hypoxia benefits MSC survival, but little is known about the underlying mechanism. This study aims to uncover potential mechanisms involving hypoxia inducible factor 1α (HIF1A) to explain the promoted MSC survival under hypoxia. MSCs were obtained from Sprague-Dawley rats and cultured under normoxia or hypoxia condition. The overexpression vector or small interfering RNA of Hif1a gene was transfected to MSCs, after which cell viability, apoptosis and expression of HIF1A were analyzed by MTT assay, flow cytometry, qRT-PCR and Western blot. Factors in p53 pathway were detected to reveal the related mechanisms. Results showed that hypoxia elevated MSCs viability and up-regulated HIF1A (P < 0.05) as previously reported. HIF1A overexpression promoted viability (P < 0.01) and suppressed apoptosis (P < 0.001) under normoxia. Correspondingly, HIF1A knockdown inhibited viability (P < 0.05) and promoted apoptosis (P < 0.01) of MSCs under hypoxia. Expression analysis suggested that p53, phosphate-p53 and p21 were repressed by HIF1A overexpression and promoted by HIF1A knockdown, and B-cell CLL/lymphoma 2 (BCL2) expression had the opposite pattern (P < 0.05). These results suggest that HIF1A may improve viability and suppress apoptosis of MSCs, implying the protective effect of HIF1A on MSC survival under hypoxia. The underlying mechanisms may involve the HIF1A-suppressed p53 pathway. This study helps to explain the mechanism of MSC survival under hypoxia, and facilitates the application of MSCs in cell therapy.
Collapse
Affiliation(s)
- Bingke Lv
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Feng Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Jie Fang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Limin Xu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Chengmei Sun
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Jianbang Han
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Tian Hua
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Zhongfei Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Zhiming Feng
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| | - Xiaodan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University; The National Key Clinical Specialty; The Engineering Technology Research Center of Education Ministry of China; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou 510282, China
| |
Collapse
|
24
|
Yang Y, Yang R, Roth M, Piperdi S, Zhang W, Dorfman H, Rao P, Park A, Tripathi S, Freeman C, Zhang Y, Sowers R, Rosenblum J, Geller D, Hoang B, Gill J, Gorlick R. Genetically transforming human osteoblasts to sarcoma: development of an osteosarcoma model. Genes Cancer 2017; 8:484-494. [PMID: 28435520 PMCID: PMC5396624 DOI: 10.18632/genesandcancer.133] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor in children and young adults. Although histologically defined by the presence of malignant osteoid, the tumor possesses lineage multipotency suggesting it could be derived from a cell anywhere on the differentiation pathway between a mesenchymal stem cell (MSC) and a mature osteoblast. To determine if preosteoblasts (pOB) could be the cell of origin differentiated MSCs were transformed with defined genetic elements. MSCs and pOB differentiated from the same MSCs were serially transformed with the oncogenes hTERT, SV40 large T antigen and H-Ras. Assays were performed to determine their tumorigenic properties, differentiation capacity and histologic appearance. When subcutaneously implanted in immunocompromised mice, cell lines derived from transformed MSC and pOB formed tumors in 4 weeks. In contrast to the transformed MSC, the pOB tumors demonstrated a histological appearance characteristic of osteosarcoma. The cell lines derived from the transformed pOB only had osteogenic and chondrogenic differentiation potential, but not adipogenic ones. However, the transformed MSC cells and standard osteosarcoma cell lines maintained their tri-lineage differentiation capacity. The inability of the transformed pOB cell line to undergo adipogenic differentiation, may suggest that osteosarcoma is derived from a cell intermediate in differentiation between an MSC and a pOB, with partial commitment to the osteoblastic lineage.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA.,Current affiliations: Department of Orthopaedic Surgery, Musculoskeletal Tumor Center, People's Hospital, Peking University, Beijing, China
| | - Rui Yang
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine of Yeshiva University and Montefiore Medical Center, Bronx, NY, USA
| | - Michael Roth
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sajida Piperdi
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wendong Zhang
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Howard Dorfman
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine of Yeshiva University and Montefiore Medical Center, Bronx, NY, USA.,Department of Pathology, Albert Einstein College of Medicine of Yeshiva University and Montefiore Medical Center, Bronx, NY, USA
| | - Pulivarthi Rao
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Amy Park
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sandeep Tripathi
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carrie Freeman
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yunjia Zhang
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rebecca Sowers
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeremy Rosenblum
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David Geller
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine of Yeshiva University and Montefiore Medical Center, Bronx, NY, USA
| | - Bang Hoang
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine of Yeshiva University and Montefiore Medical Center, Bronx, NY, USA
| | - Jonathan Gill
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Richard Gorlick
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.,Current affiliations: Pediatrics Administration, The University of Texas MD Anderson Cancer Center, Children's Cancer Hospital, Houston, TX, USA
| |
Collapse
|
25
|
Neuronal apoptosis inhibitory protein (NAIP) localizes to the cytokinetic machinery during cell division. Sci Rep 2017; 7:39981. [PMID: 28059125 PMCID: PMC5216396 DOI: 10.1038/srep39981] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 11/30/2016] [Indexed: 12/17/2022] Open
Abstract
The neuronal apoptosis inhibitory protein (NAIP) is a constituent of the inflammasome and a key component of the innate immune system. Here we use immunofluorescence to position NAIP within the cytokinetic apparatus, contiguous to chromosomal passenger complex (CPC), Centralspindlin, PRC1 and KIF4A. During metaphase, NAIP accumulates in the mitotic spindle poles and is shown in spindle microtubules; in anaphase NAIP is detected in the middle of the central spindle. At the end of cytokinesis, NAIP is localized in the outlying region of the stem body, the center of the intercellular bridge formed between daughter cells prior to cellular abscission. We also describe the sustained presence of NAIP mRNA and protein throughout the cell cycle with a significant increase observed in the G2/M phase. Consistent with a role for NAIP in cytokinesis, NAIP overexpression in HeLa cells promotes the acquisition of a multinuclear phenotype. Conversely, NAIP siRNA gene silencing results in an apoptotic lethal phenotype. Our confocal and super resolution stimulated-emission-depletion (STED) examination of mammalian cell cytokinesis demonstrate a potential new role for NAIP in addition to anti-apoptotic and innate immunology functions.
Collapse
|
26
|
Abstract
Unlike the rather stereotypic image by which it was portrayed until not too many years ago, p53 is now increasingly emerging as a multifaceted transcription factor that can sometimes exert opposing effects on biological processes. This includes pro-survival activities that seem to contradict p53's canonical proapoptotic features, as well as opposing effects on cell migration, metabolism, and differentiation. Such antagonistic bifunctionality (balancing both positive and negative signals) bestows p53 with an ideal attribute to govern homeostasis. The molecular mechanisms underpinning the paradoxical activities of p53 may be related to a protein conformational spectrum (from canonical wild-type to "pseudomutant"), diversity of DNA response elements, and/or higher-order chromatin configuration. Altogether, this functional flexibility positions p53 as a transcriptional "super hub" that dictates cell homeostasis, and ultimately cell fate, by governing a hierarchy of other functional hubs. Deciphering the mechanisms by which p53 determines which hubs to engage, and how one might modulate the preferences of p53, remains a major challenge for both basic science and translational cancer medicine.
Collapse
Affiliation(s)
- Yael Aylon
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Moshe Oren
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
27
|
Mousavinejad M, Andrews PW, Shoraki EK. Current Biosafety Considerations in Stem Cell Therapy. CELL JOURNAL 2016; 18:281-7. [PMID: 27540533 PMCID: PMC4988427 DOI: 10.22074/cellj.2016.4324] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022]
Abstract
Stem cells can be valuable model systems for drug discovery and modelling human diseases as well as to investigate cellular interactions and molecular events in the early stages of development. Controlling the differentiation of stem cells into specific germ layers provides a potential source of highly specialized cells for therapeutic applications. In recent years, finding individual properties of stem cells such as their ultimate self-renewal capacity and the generation of particular cell lines by differentiation under specific culture conditions underpins the development of regenerative therapies. These futures make stem cells a leading candidate to treat a wide range of diseases. Nevertheless, as with all novel treatments, safety issues are one of the barriers that should be overcome to guarantee the quality of a patient's life after stem cell therapy. Many studies have pointed to a large gap in our knowledge about the therapeutic applications of these cells. This gap clearly shows the importance of biosafety concerns for the current status of cell-based therapies, even more than their therapeutic efficacy. Currently, scientists report that tumorigenicity and immunogenicity are the two most important associated cell-based therapy risks. In principle, intrinsic factors such as cell characteristics and extrinsic elements introduced by manufacturing of stem cells can result in tumor formation and immunological reactions after stem cell transplantation. Therapeutic research shows there are many biological questions regarding safety issues of stem cell clinical applications. Stem cell therapy is a rapidly advancing field that needs to focus more on finding a comprehensive technology for assessing risk. A variety of risk factors (from intrinsic to extrinsic) should be considered for safe clinical stem cell therapies.
Collapse
Affiliation(s)
- Masoumeh Mousavinejad
- Centre for Stem Cell Biology (CSCB), Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | - Peter W Andrews
- Centre for Stem Cell Biology (CSCB), Department of Biomedical Science, The University of Sheffield, Sheffield, UK
| | - Elham Kargar Shoraki
- Department of Biological Sciences, Faculty of Science, Tehran Kharazmi University, Tehran, Iran
| |
Collapse
|
28
|
Lye KL, Nordin N, Vidyadaran S, Thilakavathy K. Mesenchymal stem cells: From stem cells to sarcomas. Cell Biol Int 2016; 40:610-8. [PMID: 26992453 DOI: 10.1002/cbin.10603] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/14/2016] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have garnered vast interests in clinical settings, especially in regenerative medicine due to their unique properties-they are reliably isolated and expanded from various tissue sources; they are able to differentiate into mesodermal tissues such as bones, cartilages, adipose tissues, and muscles; and they have unique immunosuppressive properties. However, there are some concerns pertaining to the role of MSCs in the human body. On one hand, they are crucial component in the regeneration and repair of the human body. On the contrary, they are shown to transform into sarcomas. Although the exact mechanisms are still unknown, many new leads have pointed to the belief that MSCs do play a role in sarcomagenesis. This review focuses on the current updates and findings of the role of MSCs in their transformation process into sarcomas.
Collapse
Affiliation(s)
- Kwan Liang Lye
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Norshariza Nordin
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Sharmili Vidyadaran
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Karuppiah Thilakavathy
- Medical Genetics Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.,Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
29
|
Pérez LM, Suárez J, Bernal A, de Lucas B, San Martin N, Gálvez BG. Obesity-driven alterations in adipose-derived stem cells are partially restored by weight loss. Obesity (Silver Spring) 2016; 24:661-9. [PMID: 26833860 DOI: 10.1002/oby.21405] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The therapeutic potential of adipose-derived stem cells (ASCs) is reduced by various stress-inducing conditions that affect tissue homeostasis such as diabetes, aging, and obesity. Previous works have provided evidence of negative effects of obesity on ASC populations, but it is unclear whether this persists after a weight loss. This study evaluated whether weight loss can restore the attenuated properties found in ASCs derived from populations with obesity (oASCs). METHODS In vitro functional analyses were performed to investigate the possible recovery properties in mouse oASCs. Using ASCs isolated from subcutaneous tissue from formerly obese mice (dASCs) and control mice (cASCs), cell proliferation, viability, and some regenerative properties in these cells were analyzed compared with oASCs to evaluate the functional cell state. RESULTS Cell proliferation, viability, and some regenerative properties are strengthened in dASCs and cASCs compared with oASCs. Nevertheless, metabolic analysis reveals a mitochondrial load misbalance and function leading to impaired respiration in dASCs. CONCLUSIONS This study demonstrates that an initial obese environment triggers a detrimental state in ASCs that is not completely recovered after weight loss.
Collapse
Affiliation(s)
- Laura M Pérez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Suárez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aurora Bernal
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz de Lucas
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Nuria San Martin
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz G Gálvez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- School of Doctorate and Research, European University, Madrid, Spain
| |
Collapse
|
30
|
Prieto C, Stam RW, Agraz-Doblas A, Ballerini P, Camos M, Castaño J, Marschalek R, Bursen A, Varela I, Bueno C, Menendez P. Activated KRAS Cooperates with MLL-AF4 to Promote Extramedullary Engraftment and Migration of Cord Blood CD34+ HSPC But Is Insufficient to Initiate Leukemia. Cancer Res 2016; 76:2478-89. [PMID: 26837759 DOI: 10.1158/0008-5472.can-15-2769] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/08/2016] [Indexed: 11/16/2022]
Abstract
The MLL-AF4 (MA4) fusion gene is the genetic hallmark of an aggressive infant pro-B-acute lymphoblastic leukemia (B-ALL). Our understanding of MA4-mediated transformation is very limited. Whole-genome sequencing studies revealed a silent mutational landscape, which contradicts the aggressive clinical outcome of this hematologic malignancy. Only RAS mutations were recurrently detected in patients and found to be associated with poorer outcome. The absence of MA4-driven B-ALL models further questions whether MA4 acts as a single oncogenic driver or requires cooperating mutations to manifest a malignant phenotype. We explored whether KRAS activation cooperates with MA4 to initiate leukemia in cord blood-derived CD34(+) hematopoietic stem/progenitor cells (HSPC). Clonogenic and differentiation/proliferation assays demonstrated that KRAS activation does not cooperate with MA4 to immortalize CD34(+) HSPCs. Intrabone marrow transplantation into immunodeficient mice further showed that MA4 and KRAS(G12V) alone or in combination enhanced hematopoietic repopulation without impairing myeloid-lymphoid differentiation, and that mutated KRAS did not cooperate with MA4 to initiate leukemia. However, KRAS activation enhanced extramedullary hematopoiesis of MA4-expressing cell lines and CD34(+) HSPCs that was associated with leukocytosis and central nervous system infiltration, both hallmarks of infant t(4;11)(+) B-ALL. Transcriptional profiling of MA4-expressing patients supported a cell migration gene signature underlying the mutant KRAS-mediated phenotype. Collectively, our findings demonstrate that KRAS affects the homeostasis of MA4-expressing HSPCs, suggesting that KRAS activation in MA4(+) B-ALL is important for tumor maintenance rather than initiation. Cancer Res; 76(8); 2478-89. ©2016 AACR.
Collapse
Affiliation(s)
- Cristina Prieto
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Ronald W Stam
- Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Antonio Agraz-Doblas
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain. Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC-CSIC-UNIVERSIDAD DE CANTABRIA-SODERCAN), Santander, Spain
| | - Paola Ballerini
- Pediatric Hematology Department, A. Trousseau Hospital, Paris, France
| | - Mireia Camos
- Hematology Laboratory, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Julio Castaño
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Rolf Marschalek
- Institute Pharmaceutical Biology, Goethe-University, Frankfurt/Main, Germany
| | - Aldeheid Bursen
- Institute Pharmaceutical Biology, Goethe-University, Frankfurt/Main, Germany
| | - Ignacio Varela
- Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC-CSIC-UNIVERSIDAD DE CANTABRIA-SODERCAN), Santander, Spain
| | - Clara Bueno
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.
| | - Pablo Menendez
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain. Instituciò Catalana Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
31
|
Velletri T, Xie N, Wang Y, Huang Y, Yang Q, Chen X, Chen Q, Shou P, Gan Y, Cao G, Melino G, Shi Y. P53 functional abnormality in mesenchymal stem cells promotes osteosarcoma development. Cell Death Dis 2016; 7:e2015. [PMID: 26775693 PMCID: PMC4816167 DOI: 10.1038/cddis.2015.367] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 11/13/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
Abstract
It has been shown that p53 has a critical role in the differentiation and functionality of various multipotent progenitor cells. P53 mutations can lead to genome instability and subsequent functional alterations and aberrant transformation of mesenchymal stem cells (MSCs). The significance of p53 in safeguarding our body from developing osteosarcoma (OS) is well recognized. During bone remodeling, p53 has a key role in negatively regulating key factors orchestrating the early stages of osteogenic differentiation of MSCs. Interestingly, changes in the p53 status can compromise bone homeostasis and affect the tumor microenvironment. This review aims to provide a unique opportunity to study the p53 function in MSCs and OS. In the context of loss of function of p53, we provide a model for two sources of OS: MSCs as progenitor cells of osteoblasts and bone tumor microenvironment components. Standing at the bone remodeling point of view, in this review we will first explain the determinant function of p53 in OS development. We will then summarize the role of p53 in monitoring MSC fidelity and in regulating MSC differentiation programs during osteogenesis. Finally, we will discuss the importance of loss of p53 function in tissue microenvironment. We expect that the information provided herein could lead to better understanding and treatment of OS.
Collapse
Affiliation(s)
- T Velletri
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - N Xie
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China.,Biochemistry Laboratory IDI-IRCC, Department of Experimental Medicine and Surgery, University of Rome Torvergata, Rome 00133, Italy
| | - Y Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Y Huang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Q Yang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - X Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Q Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - P Shou
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Y Gan
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - G Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - G Melino
- Biochemistry Laboratory IDI-IRCC, Department of Experimental Medicine and Surgery, University of Rome Torvergata, Rome 00133, Italy.,Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Y Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China.,Soochow Institutes for Translational Medicine, Soochow University, Suzhou, China
| |
Collapse
|
32
|
Sanjuan-Pla A, Romero-Moya D, Prieto C, Bueno C, Bigas A, Menendez P. Intra-Bone Marrow Transplantation Confers Superior Multilineage Engraftment of Murine Aorta-Gonad Mesonephros Cells Over Intravenous Transplantation. Stem Cells Dev 2016; 25:259-65. [PMID: 26603126 DOI: 10.1089/scd.2015.0309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Hematopoietic stem cell (HSC) engraftment has been achieved using single-cell transplantation of prospectively highly purified adult HSC populations. However, bulk transplants are still performed when assessing the HSC potential of early embryonic hematopoietic tissues such as the aorta-gonad mesonephros (AGM) due to very low HSC activity content early in development. Intra-bone marrow transplantation (IBMT) has emerged as a superior administration route over intravenous (IV) transplantation for assessing the reconstituting ability of human HSCs in the xenotransplant setting since it bypasses the requirement for homing to the BM. In this study, we compared the ability of IBMT and IV administration of embryonic day 11.5 AGM-derived cells to reconstitute the hematopoietic system of myeloablated recipients. IBMT resulted in higher levels of AGM HSC long-term multilineage engraftment in the peripheral blood, BM, spleen, and thymus of primary and secondary recipients, and in limiting dilution experiments. The administration route did not skew the multilineage contribution pattern, but IBMT conferred higher Lineage(-)Sca-1(+)c-kit(+) long-term engraftment, in line with the superior IBMT reconstitution. Therefore, IBMT represents a superior administration route to detect HSC activity from developmentally early sources with limited HSC activity content, such as the AGM.
Collapse
Affiliation(s)
- Alejandra Sanjuan-Pla
- 1 Josep Carreras Leukemia Research Institute and School of Medicine, University of Barcelona , Barcelona, Spain
| | - Damia Romero-Moya
- 1 Josep Carreras Leukemia Research Institute and School of Medicine, University of Barcelona , Barcelona, Spain
| | - Cristina Prieto
- 1 Josep Carreras Leukemia Research Institute and School of Medicine, University of Barcelona , Barcelona, Spain
| | - Clara Bueno
- 1 Josep Carreras Leukemia Research Institute and School of Medicine, University of Barcelona , Barcelona, Spain
| | - Anna Bigas
- 2 Program in Cancer Research, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) , Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Pablo Menendez
- 1 Josep Carreras Leukemia Research Institute and School of Medicine, University of Barcelona , Barcelona, Spain .,3 Institució Catalana de Recerca i Estudis Avançats (ICREA) , Barcelona, Spain
| |
Collapse
|
33
|
Chang PY, Qu YQ, Wang J, Dong LH. The potential of mesenchymal stem cells in the management of radiation enteropathy. Cell Death Dis 2015; 6:e1840. [PMID: 26247725 PMCID: PMC4558492 DOI: 10.1038/cddis.2015.189] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/02/2015] [Accepted: 06/08/2015] [Indexed: 12/20/2022]
Abstract
Although radiotherapy is effective in managing abdominal and pelvic malignant tumors, radiation enteropathy is still unavoidable. This disease severely affects the quality of life of cancer patients due to some refractory lesions, such as intestinal ischemia, mucositis, ulcer, necrosis or even perforation. Current drugs or prevailing therapies are committed to alleviating the symptoms induced by above lesions. But the efficacies achieved by these interventions are still not satisfactory, because the milieus for tissue regeneration are not distinctly improved. In recent years, regenerative therapy for radiation enteropathy by using mesenchymal stem cells is of public interests. Relevant results of preclinical and clinical studies suggest that this regenerative therapy will become an attractive tool in managing radiation enteropathy, because mesenchymal stem cells exhibit their pro-regenerative potentials for healing the injuries in both epithelium and endothelium, minimizing inflammation and protecting irradiated intestine against fibrogenesis through activating intrinsic repair actions. In spite of these encouraging results, whether mesenchymal stem cells promote tumor growth is still an issue of debate. On this basis, we will discuss the advances in anticancer therapy by using mesenchymal stem cells in this review after analyzing the pathogenesis of radiation enteropathy, introducing the advances in managing radiation enteropathy using regenerative therapy and exploring the putative actions by which mesenchymal stem cells repair intestinal injuries. At last, insights gained from the potential risks of mesenchymal stem cell-based therapy for radiation enteropathy patients may provide clinicians with an improved awareness in carrying out their studies.
Collapse
Affiliation(s)
- P-Y Chang
- 1] Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130021, China [2] Electrochemical State Key Laboratory, Changchun Institute of Applied Chemistry Academy of Science, Changchun 130021, China
| | - Y-Q Qu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130021, China
| | - J Wang
- Electrochemical State Key Laboratory, Changchun Institute of Applied Chemistry Academy of Science, Changchun 130021, China
| | - L-H Dong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
34
|
Transcriptional Dynamics of Immortalized Human Mesenchymal Stem Cells during Transformation. PLoS One 2015; 10:e0126562. [PMID: 25978455 PMCID: PMC4433180 DOI: 10.1371/journal.pone.0126562] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/03/2015] [Indexed: 12/23/2022] Open
Abstract
Comprehensive analysis of alterations in gene expression along with neoplastic transformation in human cells provides valuable information about the molecular mechanisms underlying transformation. To further address these questions, we performed whole transcriptome analysis to the human mesenchymal stem cell line, UE6E7T-3, which was immortalized with hTERT and human papillomavirus type 16 E6/E7 genes, in association with progress of transformation in these cells. At early stages of culture, UE6E7T-3 cells preferentially lost one copy of chromosome 13, as previously described; in addition, tumor suppressor genes, DNA repair genes, and apoptosis-activating genes were overexpressed. After the loss of chromosome 13, additional aneuploidy and genetic alterations that drove progressive transformation, were observed. At this stage, the cell line expressed oncogenes as well as genes related to anti-apoptotic functions, cell-cycle progression, and chromosome instability (CIN); these pro-tumorigenic changes were concomitant with a decrease in tumor suppressor gene expression. At later stages after prolong culture, the cells exhibited chromosome translocations, acquired anchorage-independent growth and tumorigenicity in nude mice, (sarcoma) and exhibited increased expression of genes encoding growth factor and DNA repair genes, and decreased expression of adhesion genes. In particular, glypican-5 (GPC5), which encodes a cell-surface proteoglycan that might be a biomarker for sarcoma, was expressed at high levels in association with transformation. Patched (Ptc1), the cell surface receptor for hedgehog (Hh) signaling, was also significantly overexpressed and co-localized with GPC5. Knockdown of GPC5 expression decreased cell proliferation, suggesting that it plays a key role in growth in U3-DT cells (transformants derived from UE6E7T-3 cells) through the Hh signaling pathway. Thus, the UE6E7T-3 cell culture model is a useful tool for assessing the functional contribution of genes showed by expression profiling to the neoplastic transformation of human fibroblasts and human mesenchymal stem cells (hMSC).
Collapse
|
35
|
Paula ACC, Martins TMM, Zonari A, Frade SPPJ, Angelo PC, Gomes DA, Goes AM. Human adipose tissue-derived stem cells cultured in xeno-free culture condition enhance c-MYC expression increasing proliferation but bypassing spontaneous cell transformation. Stem Cell Res Ther 2015; 6:76. [PMID: 25889298 PMCID: PMC4455683 DOI: 10.1186/s13287-015-0030-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/24/2015] [Accepted: 03/02/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Human adipose tissue-derived stem cells (hASCs) are attractive cells for therapeutic applications and are currently being evaluated in multiple clinical trials. Prior to their clinical application, hASCs must be expanded ex vivo to obtain the required number of cells for transplantation. Fetal bovine serum is the supplement most widely used for cell culture, but it has disadvantages and it is not safe for cell therapy due to the risks of pathogen transmission and immune reaction. Furthermore, the cell expansion poses a risk of accumulating genetic abnormalities that could lead to malignant cell transformation. In this study, our aim was to evaluate the proliferation pattern as well as the resistance to spontaneous transformation of hASCs during expansion in a xeno-free culture condition. Methods hASCs were expanded in Dulbecco’s modified Eagle’s medium supplemented with pooled allogeneic human serum or fetal bovine serum to enable a side-by-side comparison. Cell viability and differentiation capacity toward the mesenchymal lineages were assessed, along with immunophenotype. Ki-67 expression and the proliferation kinetics were investigated. The expression of the transcription factors c-FOS and c-MYC was examined with Western blot, and MYC, CDKN2A, ERBB2 and TERT gene expression was assessed with quantitative PCR. Senescence was evaluated by β-gal staining. Karyotype analysis was performed and tumorigenesis assay in vivo was also evaluated. Results The hASCs expanded in medium with pooled allogeneic human serum did not show remarkable differences in morphology, viability, differentiation capacity or immunophenotype. The main difference observed was a significantly higher proliferative effect on hASCs cultured in pooled allogeneic human serum. There was no significant difference in C-FOS expression; however, C-MYC protein expression was enhanced in pooled allogeneic human serum cultures compared to fetal bovine serum cultures. No difference was observed in MYC and TERT mRNA levels. Moreover, the hASCs presented normal karyotype undergoing senescence, and did not form in vivo tumors, eliminating the possibility that spontaneous immortalization of hASCs had occurred with pooled allogeneic human serum. Conclusions This complete characterization of hASCs cultivated in pooled allogeneic human serum, a suitable xeno-free approach, shows that pooled allogeneic human serum provides a high proliferation rate, which can be attributed for the first time to C-MYC protein expression, and showed cell stability for safe clinical applications in compliance with good manufacturing practice.
Collapse
Affiliation(s)
- Ana C C Paula
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Thaís M M Martins
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Alessandra Zonari
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Soraia P P J Frade
- Instituto Hermes Pardini, Av. das Nações, 2448, Vespasiano, Minas Gerais, 33200-000, Brazil.
| | - Patrícia C Angelo
- Instituto Hermes Pardini, Av. das Nações, 2448, Vespasiano, Minas Gerais, 33200-000, Brazil.
| | - Dawidson A Gomes
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| | - Alfredo M Goes
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, 31270-910, Brazil.
| |
Collapse
|
36
|
Diekman BO, Thakore PI, O'Connor SK, Willard VP, Brunger JM, Christoforou N, Leong KW, Gersbach CA, Guilak F. Knockdown of the cell cycle inhibitor p21 enhances cartilage formation by induced pluripotent stem cells. Tissue Eng Part A 2015; 21:1261-74. [PMID: 25517798 PMCID: PMC4394871 DOI: 10.1089/ten.tea.2014.0240] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 12/02/2014] [Indexed: 01/22/2023] Open
Abstract
The limited regenerative capacity of articular cartilage contributes to progressive joint dysfunction associated with cartilage injury or osteoarthritis. Cartilage tissue engineering seeks to provide a biological substitute for repairing damaged or diseased cartilage, but requires a cell source with the capacity for extensive expansion without loss of chondrogenic potential. In this study, we hypothesized that decreased expression of the cell cycle inhibitor p21 would enhance the proliferative and chondrogenic potential of differentiated induced pluripotent stem cells (iPSCs). Murine iPSCs were directed to differentiate toward the chondrogenic lineage with an established protocol and then engineered to express a short hairpin RNA (shRNA) to reduce the expression of p21. Cells expressing the p21 shRNA demonstrated higher proliferative potential during monolayer expansion and increased synthesis of glycosaminoglycans (GAGs) in pellet cultures. Furthermore, these cells could be expanded ∼150-fold over three additional passages without a reduction in the subsequent production of GAGs, while control cells showed reduced potential for GAG synthesis with three additional passages. In pellets from extensively passaged cells, knockdown of p21 attenuated the sharp decrease in cell number that occurred in control cells, and immunohistochemical analysis showed that p21 knockdown limited the production of type I and type X collagen while maintaining synthesis of cartilage-specific type II collagen. These findings suggest that manipulating the cell cycle can augment the monolayer expansion and preserve the chondrogenic capacity of differentiated iPSCs, providing a strategy for enhancing iPSC-based cartilage tissue engineering.
Collapse
Affiliation(s)
- Brian O. Diekman
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
| | | | - Shannon K. O'Connor
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Vincent P. Willard
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
| | - Jonathan M. Brunger
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Nicolas Christoforou
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Department of Biomedical Engineering, Khalifa University of Science, Technology and Research, Abu Dhabi, United Arab Emirates
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Charles A. Gersbach
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
37
|
Lack of activation of telomere maintenance mechanisms in human adipose stromal cells derived from fatty portion of lipoaspirates. Plast Reconstr Surg 2015; 135:114e-123e. [PMID: 25539318 DOI: 10.1097/prs.0000000000001008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Significant improvement in the understanding of mesenchymal stem cell biology paved the way to their clinical use. Human lipoaspirates derived from mesenchymal stem cells (adipose-derived stem cells) continue to draw the attention of researchers in the field of basic and applied research due to their regenerative, reparative, angiogenic, antiapoptotic, and immunosuppressive properties, all of which collectively point out their therapeutic potential. There is still, however, a need for further investigation to improve the knowledge of stem cell biology, to broaden their field of use, and to enhance their therapeutic effectiveness. METHODS The authors characterized human adipose-derived stem cells at different in vitro culture time points in terms of immunophenotype, multilineage differentiation, long-term survival with self-renewal capacity, and presence of telomere maintenance mechanisms (telomerase activity and alternative lengthening of telomere) for excluding their eventual susceptibility to malignant transformation. RESULTS Adipose-derived stem cells were isolated from the abdomen and peritrochanteric region of 31 female donors, propagated, and monitored in vitro for several passages. The outgrown cells shared the biological properties of mesenchymal stem cells, with adherence to plastic, expression of the typical surface markers, and induction of adipogenic, osteogenic, and chondrogenic differentiation. Telomerase activity and alternative lengthening of telomere mechanisms at different passages of cultures were not evidenced. CONCLUSION The results support the concept that in vitro expanded adipose-derived stem cells obtained from fat tissue are not susceptible to developing one of the hallmarks of malignant transformation and can be considered amenable for cell therapy approaches.
Collapse
|
38
|
Sharma S, Bhonde R. Influence of Nuclear Blebs and Micronuclei Status on the Growth Kinetics of Human Mesenchymal Stem Cells. J Cell Physiol 2014; 230:657-66. [DOI: 10.1002/jcp.24789] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/29/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Shikha Sharma
- School of Regenerative Medicine; Manipal University; Bangalore Karnataka India
| | - Ramesh Bhonde
- School of Regenerative Medicine; Manipal University; Bangalore Karnataka India
| |
Collapse
|
39
|
Yang J, Ren Z, Du X, Hao M, Zhou W. The role of mesenchymal stem/progenitor cells in sarcoma: update and dispute. Stem Cell Investig 2014; 1:18. [PMID: 27358864 DOI: 10.3978/j.issn.2306-9759.2014.10.01] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/10/2014] [Indexed: 12/26/2022]
Abstract
Sarcoma is the collective name for a relatively rare, yet heterogeneous group of cancers, most probably derived from mesenchymal tissues. There are currently over 50 sarcoma subtypes described underscoring the clinical and biologic diversity of this group of malignant cancers. This wide lineage range might suggest that sarcomas originate from either many committed different cell types or from a multipotent cell. Mesenchymal stem/progenitor cells (MSCs) are able to differentiate into many cell types and these multipotent cells have been isolated from several adult human tumors, making them available for research as well as potential beneficial therapeutical agents. Recent accomplishments in the field have broadened our knowledge of MSCs in relation to sarcoma origin and sarcoma treatment in therapeutic settings. However, numerous concerns and disputes have been raised about whether they are the putative originating cells of sarcoma and their questionable role in sarcomagenesis and progression. We summarize the update and dispute about MSC investigations in sarcomas including the definition, cell origin hypothesis, functional and descriptive assays, roles in sarcomagenesis and targeted therapy, with the purpose to give a comprehensive view of the role of MSCs in sarcomas.
Collapse
Affiliation(s)
- Jilong Yang
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Zhiwu Ren
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Xiaoling Du
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Mengze Hao
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| | - Wenya Zhou
- 1 Departments of Bone and Soft Tissue Tumor, 2 National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China ; 3 Departments of Diagnostics, Tianjin Medical University, Tianjin 30060, China
| |
Collapse
|
40
|
Alba-Castellón L, Batlle R, Francí C, Fernández-Aceñero MJ, Mazzolini R, Peña R, Loubat J, Alameda F, Rodríguez R, Curto J, Albanell J, Muñoz A, Bonilla F, Ignacio Casal J, Rojo F, García de Herreros A. Snail1 expression is required for sarcomagenesis. Neoplasia 2014; 16:413-21. [PMID: 24947186 PMCID: PMC4198692 DOI: 10.1016/j.neo.2014.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/15/2022] Open
Abstract
Snail1 transcriptional repressor is a major inducer of epithelial-to mesenchymal transition but is very limitedly expressed in adult animals. We have previously demonstrated that Snail1 is required for the maintenance of mesenchymal stem cells (MSCs), preventing their premature differentiation. Now, we show that Snail1 controls the tumorigenic properties of mesenchymal cells. Increased Snail1 expression provides tumorigenic capabilities to fibroblastic cells; on the contrary, Snail1 depletion decreases tumor growth. Genetic depletion of Snail1 in MSCs that are deficient in p53 tumor suppressor downregulates MSC markers and prevents the capability of these cells to originate sarcomas in immunodeficient SCID mice. Notably, an analysis of human sarcomas shows that, contrarily to epithelial tumors, these neoplasms display high Snail1 expression. This is particularly clear for undifferentiated tumors, which are associated with poor outcome. Together, our results indicate a role for Snail1 in the generation of sarcomas.
Collapse
Affiliation(s)
- Lorena Alba-Castellón
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Raquel Batlle
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Clara Francí
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | | | - Rocco Mazzolini
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Raúl Peña
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Jordina Loubat
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Francesc Alameda
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; Servei d'Anatomia Patològica, Hospital del Mar, Barcelona, Spain
| | - Rufo Rodríguez
- Departamento de Patología, Hospital Virgen de la Salud, Toledo, Spain
| | - Josué Curto
- Departament de Bioquímica i Biologia Molecular, Centre d'Estudis en Biofísica, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Joan Albanell
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; Servei d'Oncologia Mèdica, Hospital del Mar, Barcelona, Spain
| | - Alberto Muñoz
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Félix Bonilla
- Servicio de Oncología, Hospital Puerta de Hierro, Majadahonda, Spain
| | - J Ignacio Casal
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Federico Rojo
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; Servicio de Anatomía Patológica, Fundación Jiménez Díaz, Madrid, Spain
| | - Antonio García de Herreros
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
41
|
Rodriguez R, Tornin J, Suarez C, Astudillo A, Rubio R, Yauk C, Williams A, Rosu-Myles M, Funes JM, Boshoff C, Menendez P. Expression of FUS-CHOP fusion protein in immortalized/transformed human mesenchymal stem cells drives mixoid liposarcoma formation. Stem Cells 2014; 31:2061-72. [PMID: 23836491 DOI: 10.1002/stem.1472] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/22/2013] [Accepted: 06/08/2013] [Indexed: 12/11/2022]
Abstract
Increasing evidence supports that mesenchymal stromal/stem cells (MSCs) may represent the target cell for sarcoma development. Although different sarcomas have been modeled in mice upon expression of fusion oncogenes in MSCs, sarcomagenesis has not been successfully modeled in human MSCs (hMSCs). We report that FUS-CHOP, a hallmark fusion gene in mixoid liposarcoma (MLS), has an instructive role in lineage commitment, and its expression in hMSC sequentially immortalized/transformed with up to five oncogenic hits (p53 and Rb deficiency, hTERT over-expression, c-myc stabilization, and H-RAS(v12) mutation) drives the formation of serially transplantable MLS. This is the first model of sarcoma based on the expression of a sarcoma-associated fusion protein in hMSC, and allowed us to unravel the differentiation processes and signaling pathways altered in the MLS-initiating cells. This study will contribute to test novel therapeutic approaches and constitutes a proof-of-concept to use hMSCs as target cell for modeling other fusion gene-associated human sarcomas.
Collapse
Affiliation(s)
- Rene Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Shetzer Y, Kagan S, Koifman G, Sarig R, Kogan-Sakin I, Charni M, Kaufman T, Zapatka M, Molchadsky A, Rivlin N, Dinowitz N, Levin S, Landan G, Goldstein I, Goldfinger N, Pe'er D, Radlwimmer B, Lichter P, Rotter V, Aloni-Grinstein R. The onset of p53 loss of heterozygosity is differentially induced in various stem cell types and may involve the loss of either allele. Cell Death Differ 2014; 21:1419-31. [PMID: 24832469 DOI: 10.1038/cdd.2014.57] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 02/27/2014] [Accepted: 03/17/2014] [Indexed: 12/12/2022] Open
Abstract
p53 loss of heterozygosity (p53LOH) is frequently observed in Li-Fraumeni syndrome (LFS) patients who carry a mutant (Mut) p53 germ-line mutation. Here, we focused on elucidating the link between p53LOH and tumor development in stem cells (SCs). Although adult mesenchymal stem cells (MSCs) robustly underwent p53LOH, p53LOH in induced embryonic pluripotent stem cells (iPSCs) was significantly attenuated. Only SCs that underwent p53LOH induced malignant tumors in mice. These results may explain why LFS patients develop normally, yet acquire tumors in adulthood. Surprisingly, an analysis of single-cell sub-clones of iPSCs, MSCs and ex vivo bone marrow (BM) progenitors revealed that p53LOH is a bi-directional process, which may result in either the loss of wild-type (WT) or Mut p53 allele. Interestingly, most BM progenitors underwent Mutp53LOH. Our results suggest that the bi-directional p53LOH process may function as a cell-fate checkpoint. The loss of Mutp53 may be regarded as a DNA repair event leading to genome stability. Indeed, gene expression analysis of the p53LOH process revealed upregulation of a specific chromatin remodeler and a burst of DNA repair genes. However, in the case of loss of WTp53, cells are endowed with uncontrolled growth that promotes cancer.
Collapse
Affiliation(s)
- Y Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - S Kagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - G Koifman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - R Sarig
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - I Kogan-Sakin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - M Charni
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - T Kaufman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - M Zapatka
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - A Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - N Rivlin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - N Dinowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - S Levin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - G Landan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - I Goldstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - N Goldfinger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - D Pe'er
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - B Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - P Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - V Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - R Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
43
|
Mutsaers AJ, Walkley CR. Cells of origin in osteosarcoma: mesenchymal stem cells or osteoblast committed cells? Bone 2014; 62:56-63. [PMID: 24530473 DOI: 10.1016/j.bone.2014.02.003] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/14/2014] [Accepted: 02/05/2014] [Indexed: 12/14/2022]
Abstract
Osteosarcoma is a disease with many complex genetic abnormalities but few well defined genetic drivers of tumor initiation and evolution. The disease is diagnosed and defined through the observation of malignant osteoblastic cells that produce osteoid, however the exact cell of origin for this cancer remains to be definitively defined. Evidence exists to support a mesenchymal stem cell as well as committed osteoblast precursors as the cell of origin. Increasing numbers of experimental models have begun to shed light on to the likely cell population that gives rise to OS in vivo with the weight of evidence favoring an osteoblastic population as the cell of origin. As more information is gathered regarding osteosarcoma initiating cells and how they may relate to the cell of origin we will derive a better understanding of the development of this disease which may ultimately lead to clinical improvements through more personalized therapeutic approaches.
Collapse
Affiliation(s)
- Anthony J Mutsaers
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Carl R Walkley
- Stem Cell Regulation Unit and ACRF Rational Drug Discovery Centre, St. Vincent's Institute, Fitzroy, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.
| |
Collapse
|
44
|
Lucarelli E, Bellotti C, Mantelli M, Avanzini MA, Maccario R, Novara F, Arrigo G, Zuffardi O, Zuntini M, Pandolfi M, Sangiorgi L, Lisini D, Donati D, Duchi S. In vitro biosafety profile evaluation of multipotent mesenchymal stem cells derived from the bone marrow of sarcoma patients. J Transl Med 2014; 12:95. [PMID: 24716831 PMCID: PMC4022272 DOI: 10.1186/1479-5876-12-95] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/31/2014] [Indexed: 11/30/2022] Open
Abstract
Background In osteosarcoma (OS) and most Ewing sarcoma (EWS) patients, the primary tumor originates in the bone. Although tumor resection surgery is commonly used to treat these diseases, it frequently leaves massive bone defects that are particularly difficult to be treated. Due to the therapeutic potential of mesenchymal stem cells (MSCs), OS and EWS patients could benefit from an autologous MSCs-based bone reconstruction. However, safety concerns regarding the in vitro expansion of bone marrow-derived MSCs have been raised. To investigate the possible oncogenic potential of MSCs from OS or EWS patients (MSC-SAR) after expansion, this study focused on a biosafety assessment of MSC-SAR obtained after short- and long-term cultivation compared with MSCs from healthy donors (MSC-CTRL). Methods We initially characterized the morphology, immunophenotype, and differentiation multipotency of isolated MSC-SAR. MSC-SAR and MSC-CTRL were subsequently expanded under identical culture conditions. Cells at the early (P3/P4) and late (P10) passages were collected for the in vitro analyses including: sequencing of genes frequently mutated in OS and EWS, evaluation of telomerase activity, assessment of the gene expression profile and activity of major cancer pathways, cytogenetic analysis on synchronous MSCs, and molecular karyotyping using a comparative genomic hybridization (CGH) array. Results MSC-SAR displayed comparable morphology, immunophenotype, proliferation rate, differentiation potential, and telomerase activity to MSC-CTRL. Both cell types displayed signs of senescence in the late stages of culture with no relevant changes in cancer gene expression. However, cytogenetic analysis detected chromosomal anomalies in the early and late stages of MSC-SAR and MSC-CTRL after culture. Conclusions Our results demonstrated that the in vitro expansion of MSCs does not influence or favor malignant transformation since MSC-SAR were not more prone than MSC-CTRL to deleterious changes during culture. However, the presence of chromosomal aberrations supports rigorous phenotypic, functional and genetic evaluation of the biosafety of MSCs, which is important for clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Serena Duchi
- Osteoarticolar Regeneration Laboratory, Rizzoli Orthopaedic Institute, Bologna, Italy.
| |
Collapse
|
45
|
Honoki K, Tsujiuchi T. Senescence bypass in mesenchymal stem cells: a potential pathogenesis and implications of pro-senescence therapy in sarcomas. Expert Rev Anticancer Ther 2014; 13:983-96. [PMID: 23984899 DOI: 10.1586/14737140.2013.820010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cellular senescence is a mechanism that limits the lifespan of somatic cells as the results of replicative proliferation and response to stresses, and that prevents undesired oncogenic changes constituting a barrier against immortalization and tumorigenesis. Mesenchymal stem cells (MSCs) reside in a variety of tissues, and participates in tissue maintenance with their multipotent differentiation ability. MSCs are also considered to be as cells of origin for certain type of sarcomas. We reviewed the mechanisms of cellular senescence in MSCs and hypothesized senescence bypass as the potential pathogenesis for sarcoma development, and proposed the possibility of senescence induction therapy for an alternative treatment strategy against sarcomas, especially cells with the resistance to conventional chemo and radiotherapy including sarcoma stem cells.
Collapse
Affiliation(s)
- Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | |
Collapse
|
46
|
Shetzer Y, Solomon H, Koifman G, Molchadsky A, Horesh S, Rotter V. The paradigm of mutant p53-expressing cancer stem cells and drug resistance. Carcinogenesis 2014; 35:1196-208. [PMID: 24658181 DOI: 10.1093/carcin/bgu073] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is well accepted that expression of mutant p53 involves the gain of oncogenic-specific activities accentuating the malignant phenotype. Depending on the specific cancer type, mutant p53 can contribute to either the early or the late events of the multiphase process underlying the transformation of a normal cell into a cancerous one. This multifactorial system is evident in ~50% of human cancers. Mutant p53 was shown to interfere with a variety of cellular functions that lead to augmented cell survival, cellular plasticity, aberration of DNA repair machinery and other effects. All these effects culminate in the acquisition of drug resistance often seen in cancer cells. Interestingly, drug resistance has also been suggested to be associated with cancer stem cells (CSCs), which reside within growing tumors. The notion that p53 plays a regulatory role in the life of stem cells, coupled with the observations that p53 mutations may contribute to the evolvement of CSCs makes it challenging to speculate that drug resistance and cancer recurrence are mediated by CSCs expressing mutant p53.
Collapse
Affiliation(s)
- Yoav Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hilla Solomon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gabriela Koifman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alina Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Stav Horesh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
47
|
Rajamani K, Li YS, Hsieh DK, Lin SZ, Harn HJ, Chiou TW. Genetic and epigenetic instability of stem cells. Cell Transplant 2014; 23:417-33. [PMID: 24622296 DOI: 10.3727/096368914x678472] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recently, research on stem cells has been receiving an increasing amount of attention, both for its advantages and disadvantages. Genetic and epigenetic instabilities among stem cells have been a recurring obstacle to progress in regenerative medicine using stem cells. Various reports have stated that these instabilities can transform stem cells when transferred in vivo and thus have the potential to develop tumors. Previous research has shown that various extrinsic and intrinsic factors can contribute to the stability of stem cells. The extrinsic factors include growth supplements, growth factors, oxygen tension, passage technique, and cryopreservation. Controlling these factors based on previous reports may assist researchers in developing strategies for the production and clinical application of "safe" stem cells. On the other hand, the intrinsic factors can be unpredictable and uncontrollable; therefore, to ensure the successful use of stem cells in regenerative medicine, it is imperative to develop and implement appropriate strategies and technique for culturing stem cells and to confirm the genetic and epigenetic safety of these stem cells before employing them in clinical trials.
Collapse
Affiliation(s)
- Karthyayani Rajamani
- Department of Life Science and Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan
| | | | | | | | | | | |
Collapse
|
48
|
Aloni-Grinstein R, Shetzer Y, Kaufman T, Rotter V. p53: the barrier to cancer stem cell formation. FEBS Lett 2014; 588:2580-9. [PMID: 24560790 DOI: 10.1016/j.febslet.2014.02.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 02/07/2014] [Accepted: 02/07/2014] [Indexed: 02/08/2023]
Abstract
The role of p53 as the "guardian of the genome" in differentiated somatic cells, triggering various biological processes, is well established. Recent studies in the stem cell field have highlighted a profound role of p53 in stem cell biology as well. These studies, combined with basic data obtained 20 years ago, provide insight into how p53 governs the quantity and quality of various stem cells, ensuring a sufficient repertoire of normal stem cells to enable proper development, tissue regeneration and a cancer free life. In this review we address the role of p53 in genomically stable embryonic stem cells, a unique predisposed cancer stem cell model and adult stem cells, its role in the generation of induced pluripotent stem cells, as well as its role as the barrier to cancer stem cell formation.
Collapse
Affiliation(s)
- Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoav Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tom Kaufman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
49
|
Telukuntla KS, Suncion VY, Schulman IH, Hare JM. The advancing field of cell-based therapy: insights and lessons from clinical trials. J Am Heart Assoc 2013; 2:e000338. [PMID: 24113326 PMCID: PMC3835242 DOI: 10.1161/jaha.113.000338] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kartik S Telukuntla
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | | | | |
Collapse
|
50
|
p53 regulates mesenchymal stem cell-mediated tumor suppression in a tumor microenvironment through immune modulation. Oncogene 2013; 33:3830-8. [PMID: 23975435 DOI: 10.1038/onc.2013.355] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/13/2013] [Accepted: 07/24/2013] [Indexed: 01/14/2023]
Abstract
p53 is one of the most studied genes in cancer biology, and mutations in this gene may be predictive for the development of many types of cancer in humans and in animals. However, whether p53 mutations in non-tumor stromal cells can affect tumor development has received very little attention. In this study, we show that B16F0 melanoma cells form much larger tumors in p53-deficient mice than in wild-type mice, indicating a potential role of p53 deficiency in non-tumor cells of the microenvironment. As mesenchymal stem cells (MSCs) are attracted to tumors and form a major component of the tumor microenvironment, we examined the potential role of p53 status in MSCs in tumor development. We found that larger tumors resulted when B16F0 melanoma cells were co-injected with bone marrow MSCs derived from p53-deficient mice rather than MSCs from wild-type mice. Interestingly, this tumor-promoting effect by p53-deficient MSCs was not observed in non-obese diabetic/severe combined immunodeficiency mice, indicating the immune response has a critical role. Indeed, in the presence of inflammatory cytokines, p53-deficient MSCs expressed more inducible nitric oxide synthase (iNOS) and exhibited greater immunosuppressive capacity. Importantly, tumor promotion by p53-deficient MSCs was abolished by administration of S-methylisothiourea, an iNOS inhibitor. Therefore, our data demonstrate that p53 status in tumor stromal cells has a key role in tumor development by modulating immune responses.
Collapse
|