1
|
Mohanta S, Das NK, Goswami C. Taxol-treatment alters endogenous TRPV1 expression and mitochondrial membrane potential in mesenchymal stem cells: Relevant in chemotherapy-induced pathophysiology. Biochem Biophys Res Commun 2024; 737:150498. [PMID: 39128224 DOI: 10.1016/j.bbrc.2024.150498] [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: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
Microtubule-based chemotherapeutics, primarily Taxane-derived agents are still used as the major live-saving agents, yet have several side effects including serious loss of immune cells, bone density etc. which lowers the quality of life. This imposes the need to understand the effects of these agents on Mesenchymal Stem Cells (MSCs) in details. In this work we demonstrate that Taxol and Nocodazole affects the endogenous expression of TRPV1, a non-selective cation channel in MSCs. These agents also affect the status of polymerized Actin as well as Tyrosinated-tubulin, basal cytosolic Ca2+ and mitochondrial membrane potential (ΔΨm). Notably, pharmacological modulation of TRPV1 by Capsaicin or Capsazepine can also alter the above-mentioned parameters in a context-dependent manner. We suggest that endogenous expression of TRPV1 and pharmacological modulation of TRPV1 can be utilized to rescue some of these parameters effectively. These findings may have significance in the treatments and strategies with Microtubule-based chemotherapeutics and stem-cell based therapy.
Collapse
Affiliation(s)
- Sushama Mohanta
- School of Biological Sciences, National Institute of Science Education and Research, Khurda, Odisha, 752050, India; Homi Bhabha National Institute, Mumbai, 400094, India
| | - Nilesh Kumar Das
- School of Biological Sciences, National Institute of Science Education and Research, Khurda, Odisha, 752050, India; Homi Bhabha National Institute, Mumbai, 400094, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education and Research, Khurda, Odisha, 752050, India; Homi Bhabha National Institute, Mumbai, 400094, India.
| |
Collapse
|
2
|
Ataei A, Azizi M, Hajisadeghi S, Madani M, Khorami M, Hassantash S, Saeidpour Masouleh S, Barati G. The Therapeutic Effects of Mesenchymal Stem Cells and their Secretome on Oral Squamous Cell Carcinoma. Curr Mol Med 2024; 24:1195-1207. [PMID: 37366360 DOI: 10.2174/1566524023666230627151809] [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: 10/27/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
Oral cancers are prevalent in the human population, particularly in unindustrialized countries. In 90 % of oral cancers, the tumors arise from squamous cells, which is called oral squamous cell carcinoma (OSCC). Despite new treatment strategies, the morbidity and mortality rates are still high. Current treatment options including surgery, chemotherapy, and radiotherapy are not effective in the treatment of the tumor. Cell therapy with mesenchymal stem cells (MSCs) is considered one of the leading strategies in cancer treatment. However, the field of MSC therapy in OSCC is immature and ongoing studies are being conducted in experimental and pre-clinical studies. Here, we reviewed these studies to figure out whether the use of MSCs could be worthwhile in OSCC therapy or not. Both native and engineered MSCs as well as their secretome have been used in the treatment of OSCC. It seems that genetically modified MSCs or their secretome could inhibit the tumorigenesis of OSCC. However, further pre-clinical studies are required to come to a conclusion.
Collapse
Affiliation(s)
- Atefe Ataei
- Department of Periodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Azizi
- Department of Periodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Samira Hajisadeghi
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Qom University of Medical Sciences, Qom, Iran
| | - Mojan Madani
- Orthodontics Department, Dental Faculty, Arak UNDUniversity of Medical Sciences, Arak, Iran
| | - Mozhgan Khorami
- Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Hassantash
- Faculty of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ghasem Barati
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Stem Cell Technology Research Center, Tehran, Iran
| |
Collapse
|
3
|
Katzerke C, Schaffrath J, Lützkendorf J, Janssen M, Merbach AK, Nerger K, Binder M, Baum C, Lauer K, Rohde C, Willscher E, Müller-Tidow C, Müller LP. Reduced proliferation of bone marrow MSC after allogeneic stem cell transplantation is associated with clinical outcome. Blood Adv 2023; 7:2811-2824. [PMID: 36763527 PMCID: PMC10279553 DOI: 10.1182/bloodadvances.2022008510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/30/2022] [Accepted: 01/02/2023] [Indexed: 02/11/2023] Open
Abstract
Engraftment and differentiation of donor hematopoietic stem cells is decisive for the clinical success of allogeneic stem cell transplantation (alloSCT) and depends on the recipient's bone marrow (BM) niche. A damaged niche contributes to poor graft function after alloSCT; however, the underlying mechanisms and the role of BM multipotent mesenchymal stromal cells (MSC) are ill-defined. Upon multivariate analysis in 732 individuals, we observed a reduced presence of proliferation-capable MSC in BM aspirates from patients (N = 196) who had undergone alloSCT. This was confirmed by paired analysis in 30 patients showing a higher frequency of samples with a lack of MSC presence post-alloSCT compared with pre-alloSCT. This reduced MSC presence was associated with reduced survival of patients after alloSCT and specifically with impaired graft function. Post-alloSCT MSC showed diminished in vitro proliferation along with a transcriptional antiproliferative signature, upregulation of epithelial-mesenchymal transition and extracellular matrix pathways, and altered impact on cytokine release upon contact with hematopoietic cells. To avoid in vitro culture bias, we isolated the CD146+/CD45-/HLA-DR- BM cell fraction, which comprised the entire MSC population. The post-alloSCT isolated native CD146+MSC showed a similar reduction in proliferation capacity and shared the same antiproliferative transcriptomic signature as for post-alloSCT colony-forming unit fibroblast-derived MSC. Taken together, our data show that alloSCT confers damage to the proliferative capacity of native MSC, which is associated with reduced patient survival after alloSCT and impaired engraftment of allogeneic hematopoiesis. These data represent the basis to elucidate mechanisms of BM niche reconstitution after alloSCT and its therapeutic manipulation.
Collapse
Affiliation(s)
- Christiane Katzerke
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Judith Schaffrath
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Jana Lützkendorf
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Maike Janssen
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Anne-Kathrin Merbach
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Katrin Nerger
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Mascha Binder
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Cornelia Baum
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Kirstin Lauer
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Christian Rohde
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Edith Willscher
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| | - Carsten Müller-Tidow
- Klinik für Innere Medizin V, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory-Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Lutz P. Müller
- Universitätsklinik und Poliklinik für Innere Medizin IV, Universitätsklinikum Halle, Halle (Saale), Germany
| |
Collapse
|
4
|
Večerić-Haler Ž, Sever M, Kojc N, Halloran PF, Boštjančič E, Mlinšek G, Oblak M, Poženel P, Švajger U, Hartman K, Kneževič M, Barlič A, Girandon L, Aleš Rigler A, Zver S, Buturović Ponikvar J, Arnol M. Autologous Mesenchymal Stem Cells for Treatment of Chronic Active Antibody-Mediated Kidney Graft Rejection: Report of the Phase I/II Clinical Trial Case Series. Transpl Int 2022; 35:10772. [PMID: 36484064 PMCID: PMC9722440 DOI: 10.3389/ti.2022.10772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022]
Abstract
Mesenchymal stem cell (MSCs) therapy has already been studied in kidney transplant recipients (KTRs), and the available data showed that it is safe and well tolerated. The aim of this study was to evaluate the safety and efficacy of autologous MSCs in combination with standard therapy in KTRs with biopsy-proven chronic active antibody-mediated rejection (AMR). Patients with biopsy-proven chronic active AMR received treatment with autologous bone marrow-derived MSCs (3 × 106 cells/kg iv) after completion of standard therapy and were followed for up to 12 months. The primary endpoints were safety by assessment of adverse events. Secondary endpoints included assessment of kidney graft function, immunological and histological changes related to AMR activity and chronicity assessed by conventional microscopy and molecular transcripts. A total of 3 patients were enrolled in the study before it was terminated prematurely because of adverse events. We found that AMR did not improve in any of the patients after treatment with MSCs. In addition, serious adverse events were observed in one case when autologous MSCs therapy was administered in the late phase after kidney transplantation, which requires further elucidation.
Collapse
Affiliation(s)
- Željka Večerić-Haler
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,*Correspondence: Željka Večerić-Haler,
| | - Matjaž Sever
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Nika Kojc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Philip F. Halloran
- Division of Nephrology and Transplant Immunology, Alberta Transplant Applied Genomics Centre, University of Alberta, Edmonton, AB, Canada
| | - Emanuela Boštjančič
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Mlinšek
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Manca Oblak
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Primož Poženel
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Urban Švajger
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Katrina Hartman
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | | | - Ariana Barlič
- Educell d.o.o Cell Therapy Service, Ljubljana, Slovenia
| | | | - Andreja Aleš Rigler
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Samo Zver
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jadranka Buturović Ponikvar
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Arnol
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
5
|
Kitaeva KV, Chulpanova DS, Zhuravleva MN, Filin IY, Deviatiiarov RM, Ballard-Reisch AC, Rizvanov AA, Solovyeva VV. Characteristics and Resistance to Cisplatin of Human Neuroblastoma Cells Co-Cultivated with Immune and Stromal Cells. Bioengineering (Basel) 2022; 9:655. [PMID: 36354566 PMCID: PMC9687654 DOI: 10.3390/bioengineering9110655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/21/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2023] Open
Abstract
We investigated the features of the morphology and cytokine profiles of neuroblastoma SH-SY5Y cells, bone marrow-derived mesenchymal stromal/stem cells (BM-MSCs), and peripheral blood mononuclear cells (PBMCs) in double (BM-MSCs + SH-SY5Y cells) and triple (BM-MSCs + SH-SY5Y cells + PBMCs) co-cultures incubated on plastic and Matrigel. Cells in the co-cultures communicated by vesicular transport and by exchanging membrane and cytoplasmic components. The cytokine profile of double and triple co-cultures incubated on Matrigel and plastic had differences and showed the highest concentration of a number of chemokines/cytokines, such as CXCL8/IL-8, I-TAC/CXCL11, IP10/CXCL10, MDC/CCL22, MIP-1α/CCL3, IL-1β, ENA-78/CXCL5, Gro-α/CXCL1, MCP-1/CCL2, TERC/CCL25, CXCL8/IL-8, and IL-6. High concentrations of inflammatory chemokines/cytokines in the conditioned medium of triple co-culture form a chronic inflammation, which brings the presented co-cultivation system closer to a natural tumor. Triple co-cultures were more resistant to cisplatin (CDDP) than the double- and monoculture of SH-SY5Y. The mRNA levels of BCL2, BCL2L1, RAC1, CAV1, CASP3, and BAX genes were changed in cells after co-culturing and CDDP treatment in double and triple co-cultures. The expression of the BCL2, BAX, CAV1, and CASP3 proteins in SH-SY5Y cells after the triple co-culture and CAV1 and BAX protein expression in SH-SY5Y cells after the double co-culture were determined. This study demonstrated the nature of the cellular interactions between components of tumor niche and the intercellular influence on chemoresistance observed in our tumor model, which should enable the development of novel test systems for anti-tumor agents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
6
|
Comparison of Different Clinical Chemotherapeutical Agents’ Toxicity and Cell Response on Mesenchymal Stem Cells and Cancer Cells. Cells 2022; 11:cells11192942. [PMID: 36230904 PMCID: PMC9563435 DOI: 10.3390/cells11192942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/08/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) or fibroblasts are one of the most abundant cell types in the tumor microenvironment (TME) exerting various anti- and pro-apoptotic effects during tumorigenesis, invasion, and drug treatment. Despite the recently discovered importance of MSCs in tumor progression and therapy, the response of these cells to chemotherapeutics compared to cancer cells is rarely investigated. A widely accepted view is that these naive MSCs have higher drug tolerance than cancer cells due to a significantly lower proliferation rate. Here, we examine the differences and similarities in the sensitivity of MSCs and cancer cells to nine diverse chemotherapy agents and show that, although MSCs have a slower cell cycle, these cells are still sensitive to various drugs. Surprisingly, MSCs showed similar sensitivity to a panel of compounds, however, suffered fewer DNA double-stranded breaks, did not enter into a senescent state, and was virtually incapable of apoptosis. Our results suggest that MSCs and cancer cells have different cell fates after drug treatment, and this could influence therapy outcome. These findings could help design drug combinations targeting both MSCs and cancer cells in the TME.
Collapse
|
7
|
Santos A, Domingues C, Jarak I, Veiga F, Figueiras A. Osteosarcoma from the unknown to the use of exosomes as a versatile and dynamic therapeutic approach. Eur J Pharm Biopharm 2021; 170:91-111. [PMID: 34896571 DOI: 10.1016/j.ejpb.2021.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 12/18/2022]
Abstract
The most common primary malignant tumor of bone in children is osteosarcoma (OS). Nowadays, the prognosis and the introduction of chemotherapy in OS have improved survival rates of patients. Nevertheless, the results are still unsatisfactory, especially, in patients with recurrent disease or metastatic. OS chemotherapy has two main challenges related to treatment toxicity and multiple drug resistance. In this way, nanotechnology has developed nanosystems capable of releasing the drug directly at the OS cells and decreasing the drug's toxicity. Exosomes (Exo), a cell-derived nano-sized and a phospholipid vehicle, have been recognized as important drug delivery systems in several cancers. They are involved in a variety of biological processes and are an important mediator of long-distance intercellular communication. Exo can reduce inflammation and show low toxicity in healthy cells. Furthermore, the incorporation of specific proteins or peptides on the Exo surface improves their targeting capability in several clinical applications. Due to their unique structure and relevant characteristics, Exo is a promising nanocarrier for OS treatment. This review intends to describe the properties that turn Exo into an efficient, as well as safe nanovesicle for drug delivery and treatment of OS.
Collapse
Affiliation(s)
- Ana Santos
- Univ Coimbra, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, Coimbra, Portugal
| | - Cátia Domingues
- Univ Coimbra, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, LAQV, REQUIMTE, Faculty of Pharmacy, Portugal; Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
| | - Ivana Jarak
- Univ Coimbra, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, Coimbra, Portugal
| | - Francisco Veiga
- Univ Coimbra, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, LAQV, REQUIMTE, Faculty of Pharmacy, Portugal
| | - Ana Figueiras
- Univ Coimbra, Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, LAQV, REQUIMTE, Faculty of Pharmacy, Portugal.
| |
Collapse
|
8
|
Platoff R, Villalobos MA, Hagaman AR, Liu Y, Matthews M, DiSanto ME, Carpenter JP, Zhang P. Effects of radiation and chemotherapy on adipose stem cells: Implications for use in fat grafting in cancer patients. World J Stem Cells 2021; 13:1084-1093. [PMID: 34567427 PMCID: PMC8422936 DOI: 10.4252/wjsc.v13.i8.1084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/30/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Autologous fat transplantation is a versatile tool in reconstructive surgery. Adipose-derived stem cells (ASCs) increase survival of fat grafts and thus are increasingly used for breast reconstruction in breast cancer patients. However, radiation and/or chemotherapy have been proposed to inhibit soft tissue regeneration in wound healing thus suggesting alteration in stem cell pathways. Therefore, elucidating effects of radiation and chemotherapy on ASCs is critical if one desires to enhance the survival of fat grafts in patients. This review outlines our work evaluating the function and recoverability of ASCs from radiation or chemotherapy patients, focusing specifically on their availability as a source of autologous stem cells for fat grafting and breast reconstruction in cancer patients. Even though evidence suggests radiation and chemotherapy negatively influence ASCs at the cellular level, the efficiency of the isolation and differentiation capacity did not appear influenced in patients after receiving chemotherapy treatment, although fat from radiated patients exhibited significantly altered ASC differentiation into endothelial-like cells. Further, the in vitro growth rates of patient’s ASCs do not differ significantly before or after treatment. Taken together, these studies suggest ASCs as an important new tool for grafting and reconstruction even when radiation and chemotherapy treatment are involved.
Collapse
Affiliation(s)
- Rebecca Platoff
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
| | - Miguel A Villalobos
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
| | - Ashleigh Rapp Hagaman
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
| | - Yuan Liu
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Martha Matthews
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Michael E DiSanto
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Jeffrey P Carpenter
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Ping Zhang
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, United States
- Department of Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| |
Collapse
|
9
|
Synergistic Effect of the Long-Term Overexpression of Bcl-2 and BDNF Lentiviral in Cell Protecting against Death and Generating TH Positive and CHAT Positive Cells from MSC. Int J Mol Sci 2021; 22:ijms22137086. [PMID: 34209365 PMCID: PMC8268627 DOI: 10.3390/ijms22137086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSC) are potentially a good material for transplantation in many diseases, including neurodegenerative diseases. The main problem with using them is the low percentage of surviving cells after the transplant procedure and the naturally poor ability of MSC to spontaneously differentiate into certain types of cells, which results in their poor integration with the host cells. The aim and the novelty of this work consists in the synergistic overexpression of two genes, BCL2 and BDNF, using lentiviral vectors. According to our hypothesis, the overexpression of the BCL2 gene is aimed at increasing the resistance of cells to stressors and toxic factors. In turn, the overexpression of the BDNF gene is suspected to direct the MSC into the neural differentiation pathway. As a result, it was shown that the overexpression of both genes and the overproduction of proteins is permanent and persists for at least 60 days. The synergistically transduced MSC were significantly more resistant to the action of staurosporine; 12 days after transduction, the synergistically transduced MSC had a six-times greater survival rate. The overexpression of the Bcl-2 and BDNF proteins was sufficient to stimulate a significant overexpression of the CHAT gene, and under specific conditions, the TH, TPH1, and SYP genes were also overexpressed. Modified MSC are able to differentiate into cholinergic and dopaminergic neurons, and the release of acetylcholine and dopamine may indicate their functionality.
Collapse
|
10
|
Zohni K, Lopez L, Mander P, Szaraz P, Filice M, Wyse BA, Garcia M, Gat I, Glass K, Gauthier-Fisher A, Librach CL. Human umbilical cord perivascular cells maintain regenerative traits following exposure to cyclophosphamide. Cancer Lett 2020; 501:133-146. [PMID: 33387641 DOI: 10.1016/j.canlet.2020.12.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 11/19/2022]
Abstract
Chemotherapies can cause germ cell depletion and gonadal failure. When injected post-chemotherapy, mesenchymal stromal cells (MSCs) from various sources have been shown to have regenerative effects in rodent models of chemotherapy-induced gonadal injury. Here, we evaluated two properties of a novel source of MSC, first trimester (FTM) human umbilical cord perivascular cells (HUCPVCs) (with increased regenerative potential compared to older sources), that may render them a promising candidate for chemotherapeutic gonadal injury prevention. Firstly, their ability to resist the cytotoxic effects of cyclophosphamide (CTX) in vitro, as compared to term HUCPVCs and bone marrow cells (BMSCs); and secondly, whether they prevent gonadal dysfunction if delivered prior to gonadotoxic therapy in vivo. BMSC, FTM HUCPVC, term HUCPVC, and control NTERA2 cells were treated with moderate (150 μmol/L) and high (300 μmol/L) doses of CTX in vitro. Viability, proliferative capacity, mesenchymal cell lineage markers and differentiation capacity, immunogenicity, and paracrine gene expression were assessed. CTX was administered to Wistar rats 2 days following an intra-ovarian injection of FTM HUCPVC. HUCPVC survival and ovarian follicle numbers were assessed using histological methods. We conclude that FTM HUCPVC maintain key regenerative properties following chemotherapy exposure and that pre-treatment with these cells may prevent CTX-induced ovarian damage in vivo. Therefore, HUCPVCs are promising candidates for fertility preservation.
Collapse
Affiliation(s)
- Khaled Zohni
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics & Gynecology, University of Toronto, Toronto, Canada; Department of Obstetrics and Gynecology, University of Manitoba, Winnipeg, Canada; Heartland Fertility and Gynecology Clinic, Winnipeg, Manitoba, Canada
| | - Lianet Lopez
- CReATe Fertility Centre, Toronto, Ontario, Canada
| | | | - Peter Szaraz
- CReATe Fertility Centre, Toronto, Ontario, Canada
| | | | | | | | - Itai Gat
- CReATe Fertility Centre, Toronto, Ontario, Canada; Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Tel HaShomer, Ramat Gan, Affiliated to Sackler Medical School, University of Tel Aviv, Israel
| | - Karen Glass
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics & Gynecology, University of Toronto, Toronto, Canada
| | | | - Clifford L Librach
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics & Gynecology, University of Toronto, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada; Department of Gynecology, Women's College Hospital, Toronto, ON, Canada.
| |
Collapse
|
11
|
The Therapeutic Potential of Mesenchymal Stromal Cells in the Treatment of Chemotherapy-Induced Tissue Damage. Stem Cell Rev Rep 2020; 15:356-373. [PMID: 30937640 DOI: 10.1007/s12015-019-09886-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chemotherapy constitutes one of the key treatment modalities for solid and hematological malignancies. Albeit being an effective treatment, chemotherapy application is often limited by its damage to healthy tissues, and curative treatment options for chemotherapy-related side effects are largely missing. As mesenchymal stromal cells (MSCs) are known to exhibit regenerative capacity mainly by supporting a beneficial microenvironment for tissue repair, MSC-based therapies may attenuate chemotherapy-induced tissue injuries. An increasing number of animal studies shows favorable effects of MSC-based treatments; however, clinical trials for MSC therapies in the context of chemotherapy-related side effects are rare. In this concise review, we summarize the current knowledge of the effects of MSCs on chemotherapy-induced tissue toxicities. Both preclinical and early clinical trials investigating MSC-based treatments for chemotherapy-related side reactions are presented, and mechanistic explanations about the regenerative effects of MSCs in the context of chemotherapy-induced tissue damage are discussed. Furthermore, challenges of MSC-based treatments are outlined that need closer investigations before these multipotent cells can be safely applied to cancer patients. As any pro-tumorigenicity of MSCs needs to be ruled out prior to clinical utilization of these cells for cancer patients, the pro- and anti-tumorigenic activities of MSCs are discussed in detail.
Collapse
|
12
|
Hagaman AR, Zhang P, Koko KR, Nolan RS, Fromer MW, Gaughan J, Matthews M. Isolation and identification of adipose-derived stromal/stem cells from breast cancer patients after exposure neoadjuvant chemotherapy. World J Exp Med 2020; 10:26-40. [PMID: 32399395 PMCID: PMC7203539 DOI: 10.5493/wjem.v10.i3.26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/04/2020] [Accepted: 03/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND With recent research advances, adipose-derived stromal/stem cells (ASCs) have been demonstrated to facilitate the survival of fat grafts and thus are increasingly used for reconstructive procedures following surgery for breast cancer. Unfortunately, in patients, following radiation and chemotherapy for breast cancer suggest that these cancer treatment therapies may limit stem cell cellular functions important for soft tissue wound healing. For clinical translation to patients that have undergone cancer treatment, it is necessary to understand the effects of these therapies on the ASC's ability to improve fat graft survival in clinical practice. AIM To investigate whether the impact on ASCs function capacity and recovery in cancer patients may be due to the chemotherapy. METHODS ASCs were isolated from the cancerous side and noncancerous side of the breast from the same patients with receiving neoadjuvant chemotherapy (NAC) or not-receiving NAC. ASCs were in vitro treated with 5-fluorouracil (5-FU), doxorubicin (DXR), and cyclophosphamide (Cytoxan) at various concentrations. The stem cells yield, cell viability, and proliferation rates were measured by growth curves and MTT assays. Differentiation capacity for adipogenesis was determined by qPCR analysis of the specific gene markers and histological staining. RESULTS No significant differences were observed between the yield of ASCs in patients receiving NAC treatment and not-receiving NAC. ASCs yield from the cancerous side of the breast showed lower than the noncancerous side of the breast in both patients receiving NAC and not-receiving NAC. The proliferation rates of ASCs from patients didn't differ much before and after NAC upon in vitro culture, and these cells appeared to retain the capacity to acquire adipocyte traits simile to the ASCs from patients not-receiving NAC. After cessation and washout of the drugs for another a week of culturing, ASCs showed a slow recovery of cell growth capacity in 5-FU-treated groups but was not observed in ASCs treated with DXR groups. CONCLUSION Neoadjuvant therapies do not affect the functioning capacity of ASCs. ASCs may hold great potential to serve as a cell source for fat grafting and reconstruction in patients undergoing chemotherapy.
Collapse
Affiliation(s)
| | - Ping Zhang
- Department of Surgery, Cooper University Hospital, Camden, NJ 08103, United States
- Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Kiavash R Koko
- Department of Surgery, Cooper University Hospital, Camden, NJ 08103, United States
| | - Ryan S Nolan
- Department of Surgery, Cooper University Hospital, Camden, NJ 08103, United States
| | - Marc W Fromer
- Department of Surgery, Cooper University Hospital, Camden, NJ 08103, United States
| | - John Gaughan
- Department of Surgery, Cooper University Hospital, Camden, NJ 08103, United States
- Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - Martha Matthews
- Department of Surgery, Cooper University Hospital, Camden, NJ 08103, United States
- Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| |
Collapse
|
13
|
Black L, Zorina T. Cell-based immunomodulatory therapy approaches for type 1 diabetes mellitus. Drug Discov Today 2020; 25:380-391. [DOI: 10.1016/j.drudis.2019.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/11/2019] [Accepted: 11/30/2019] [Indexed: 12/14/2022]
|
14
|
Yu S, Mu Y, Zhang X, Li J, Lee C, Wang H. Molecular mechanisms underlying titanium dioxide nanoparticles (TiO 2NP) induced autophagy in mesenchymal stem cells (MSC). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:997-1008. [PMID: 31718501 DOI: 10.1080/15287394.2019.1688482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The bone marrow is one of the target tissues for titanium dioxide nanoparticles (TiO2NP) following environmental exposure. At present, the consequences of TiO2NP exposure in bone are not well known. The aim of this study was to investigate the effects of TiO2NP on mesenchymal stem cells (MSCs) and potential underlying mechanisms. Mesenchymal bone marrow-derived cells were cultured and treated with various concentrations of TiO2NP. Results showed that TiO2NP incubation produced cytotoxicity as evidenced by reduced cell viability. Using Western blotting TiO2NP was found to increase autophagy as determined by elevation in ratio of LC3-II from LC3-I without evidence of necrotic cell death as estimated by lactic dehydrogenase (LDH) level. TiO2NP produced a rise in intracellular reactive oxygen species (ROS) levels. The observed alterations in autophagy and oxidant stress were associated with upregulation of protein expression of p38, JNK, and ERK. Data indicate that TiO2NP-mediated decrease in MSC survival involves a complex series of events associated stimulation of mitogen-activated protein kinase (MAPK) pathway and consequent autophagy and oxidative damage.
Collapse
Affiliation(s)
- Shunbang Yu
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
| | - Yongping Mu
- Tumor Molecular Diagnostic Laboratory, Department of Clinical Laboratory Centre, The Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xudong Zhang
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
| | - Jian Li
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, US
| | - Charles Lee
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Singapore
| | - He Wang
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
| |
Collapse
|
15
|
Mesenchymal stem cells preserve their stem cell traits after exposure to antimetabolite chemotherapy. Stem Cell Res 2019; 40:101536. [PMID: 31437767 DOI: 10.1016/j.scr.2019.101536] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/29/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
|
16
|
Bajelan B, Zaki-Dizaji M, Rahmani B, Darzi S, Darabi S, Rajaei F. Resistance of human primary mesenchymal stem cells to cytotoxic effects of nutlin-3 in vitro. J Cell Biochem 2019; 121:788-796. [PMID: 31452266 DOI: 10.1002/jcb.29324] [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: 11/11/2018] [Accepted: 01/28/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND The small-molecule nutlin-3 was found to be an effective therapeutic compound and p53 activator, and acts as a murine double minute 2 antagonist, although these findings need to be clinically confirmed. The essential components of the bone marrow include mesenchymal stem cells (MSCs), which play a key role in protecting, regenerating, and proliferating hematopoietic stem cells (HSCs). This feature is vital for HSC after exposure to myelotoxic anticancer agents; nevertheless, the effects of nutlin-3 on MSCs remain to be disclosed. The present research study was conducted to examine the antiproliferative and proapoptotic effectiveness of nutlin-3 in bone marrow MSCs (BMSCs). MATERIALS AND METHODS Human-derived BMSCs were cultured for different durations, that is, 24, 48, and 72 hours, and treated using various concentrations of nutlin-3, including 5, 10, 25, 50, and 100 μΜ. To investigate the effect of nutlin-3 on the apoptosis, cell vitality and proliferation in BMSCs, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), thiazolyl blue tetrazolium bromide, propidium iodide (PI) and annexin V assay, as well as real-time polymerase chain reaction, were used. RESULTS BMSCs viability significantly decreased (P < .05) in the cells treated at concentrations of 50 and 100 μM for 24 hours and concentrations of 25, 50, and 100 μM for 48 hours and at all concentrations for 72 hours. The apoptosis of BMSCs (TUNEL positive) was significantly more visible at concentrations of 25 and 50 μM compared with that in the controls (P < .05), while this increased through dose-dependent processes. Annexin V/PI staining revealed negligible dose-dependent increases in all the apoptotic cells after 72 hours of incubation, and this apoptosis elevation was significant at 25 and 50 μM (P < .05). CONCLUSION Resistance to nutlin-3 was observed in human bone marrow-derived MSCs; nevertheless, further clinical data are required to be obtained with long-duration exposure to confirm the present findings.
Collapse
Affiliation(s)
- Babak Bajelan
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Majid Zaki-Dizaji
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Rahmani
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sina Darzi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
17
|
Werner S, Lützkendorf J, Müller T, Müller LP, Posern G. MRTF-A controls myofibroblastic differentiation of human multipotent stromal cells and their tumour-supporting function in xenograft models. Sci Rep 2019; 9:11725. [PMID: 31409840 PMCID: PMC6692381 DOI: 10.1038/s41598-019-48142-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Tumour growth and metastatic colonization is strongly influenced by the tumour stroma, including cancer-associated fibroblasts (CAF). Multipotent mesenchymal stromal cells (MSC) are a possible source of CAF following myofibroblastic differentiation, and we have previously shown that MSC support tumour growth. Triggered by tumour cell-derived factors like transforming growth factor β1 (TGF-β1), myofibroblastic MSC differentiation is associated with the increased expression of markers including alpha smooth muscle actin (α-SMA). Here we show that myocardin-related transcription factor A (MRTF-A) plays an important role in myofibroblastic differentiation of primary human MSC in vitro and their tumour-supporting function in vivo. Recombinant TGF-β1 or tumour cell conditioned medium (TCM) elevated α-SMA, calponin 1 and collagen 1 A1 (COL1A1) amount on mRNA and protein level in MSC. This correlated with increased MRTF-A activity during MSC differentiation. MRTF-A knockdown by siRNA or shRNA impaired TGF-β1 and TCM induction of α-SMA and calponin 1, but not of COL1A1. Mixed xenograft experiments using HCT8 colorectal carcinoma cells and primary MSC of different donors revealed a significant reduction in tumour weight and volume upon MRTF-A knockdown in MSC. Our study suggests that MRTF-A is involved in the functional differentiation of MSC towards a tumour-promoting CAF phenotype in vivo.
Collapse
Affiliation(s)
- Sara Werner
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Jana Lützkendorf
- University Clinic of Internal Medicine IV, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Thomas Müller
- University Clinic of Internal Medicine IV, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Lutz P Müller
- University Clinic of Internal Medicine IV, Medical Faculty, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany.
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany.
| |
Collapse
|
18
|
Theodorakos I, Paterakis G, Papadakis V, Vicha A, Topakas G, Jencova P, Karchilaki E, Taparkou A, Tsagarakis NJ, Polychronopoulou S. Interference of bone marrow CD56 + mesenchymal stromal cells in minimal residual disease investigation of neuroblastoma and other CD45 - /CD56 + pediatric malignancies using flow cytometry. Pediatr Blood Cancer 2019; 66:e27799. [PMID: 31066205 DOI: 10.1002/pbc.27799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bone marrow (BM) samples obtained from minimal residual disease (MRD)-negative children with B-cell acute lymphoblastic leukemia (B-ALL) were used in our laboratory as negative biological controls for the development of a neuroblastoma (NBL) flow-cytometric (FC) protocol. The accidental, but systematic, identification of rare cell populations (RCP) mimicking NBL cells (CD45- /CD56+ ) in these samples indicated the need for their thorough immunophenotypic identification, in order to elucidate their possible interference in NBL-MRD assessment. PROCEDURE RCP observed in BM samples from 14 children recovering from BM aplasia due to intensive chemotherapy for B-ALL were investigated with the following markers: CD81, CD200, CD24, GD2, CD73, CD13, CD90, CD146, CD9, CD117, CD10, CD99, and NG2. BM samples from six newly diagnosed patients with NBL and an NBL cell line were simultaneously investigated as positive controls. RESULTS The frequency of RCP in B-ALL BM samples was < 1/1 × 104 cells (bulky lysis), and their immunophenotypic profile was indicative of CD56+ mesenchymal stromal cells (MSCs) (CD45- , CD90+ , CD146+ , CD73+ ). Also, RCP expressed CD81 and CD200, simulating NBL cells. The most useful discriminative markers for CD56+ MSCs were CD13 and CD73. An appropriate protocol consisting of two tubes with seven color combinations was further proposed: SYTO-16, GD2 (first tube) or CD73 (second tube)-PE, CD24-ECD, CD13-PC5.5, CD45-PC7, CD81-APC, and CD56-APC700. CONCLUSIONS RCP that were immunophenotypically similar to NBL were identified as CD56+ MSCs. As these cells might pose an obstacle to accurate NBL disease assessment by FC, especially MRD, an enhanced NBL-FC protocol is proposed for prospective evaluation.
Collapse
Affiliation(s)
- Ioannis Theodorakos
- Flow Cytometry Laboratory, Department of Immunology, Athens Regional General Hospital "G. Gennimatas,", Athens, Greece
| | - Georgios Paterakis
- Flow Cytometry Laboratory, Department of Immunology, Athens Regional General Hospital "G. Gennimatas,", Athens, Greece
| | - Vassilios Papadakis
- Department of Pediatric Hematology-Oncology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Ales Vicha
- Department of Pediatric Hematology and Oncology, Charles University, 2nd Faculty of Medicine and Faculty Hospital Motol, Prague, Czech Republic
| | - Georgios Topakas
- Flow Cytometry Laboratory, Department of Immunology, Athens Regional General Hospital "G. Gennimatas,", Athens, Greece
| | - Pavla Jencova
- Department of Pediatric Hematology and Oncology, Charles University, 2nd Faculty of Medicine and Faculty Hospital Motol, Prague, Czech Republic
| | - Eirini Karchilaki
- Flow Cytometry Laboratory, Department of Immunology, Athens Regional General Hospital "G. Gennimatas,", Athens, Greece
| | - Anna Taparkou
- 1st Department of Pediatrics, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Nikolaos J Tsagarakis
- Flow Cytometry Laboratory, Department of Immunology, Athens Regional General Hospital "G. Gennimatas,", Athens, Greece
| | - Sophia Polychronopoulou
- Department of Pediatric Hematology-Oncology, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
19
|
Wu PK, Wang JY, Chen CF, Chao KY, Chang MC, Chen WM, Hung SC. Early Passage Mesenchymal Stem Cells Display Decreased Radiosensitivity and Increased DNA Repair Activity. Stem Cells Transl Med 2019; 6:1504-1514. [PMID: 28544661 PMCID: PMC5689774 DOI: 10.1002/sctm.15-0394] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 12/21/2016] [Indexed: 12/24/2022] Open
Abstract
Cell therapies using human mesenchymal stem cells (MSCs) have received much attention in the past decade. In pursuit of the therapeutic potential of MSCs, cell expansion is required to generate a great number of cells with desired phenotype and functionality. Long‐term expansion in vitro, however, can lead to altered functions. To explore the changes in DNA damage responses (DDR) in MSCs expanded, DDR pathways following irradiation were characterized in early‐ and late‐passage bone marrow MSCs. Seventy‐two hours after irradiation, the percentage of sub‐G1 cells in early‐passage MSCs did not change significantly. Reduced TUNEL staining was observed in early‐passage MSCs compared to late‐passage MSCs 4 h after irradiation. Comet assay also revealed that early‐passage MSCs were more resistant to irradiation or DNA damages induced by genotoxic agents than late‐passage MSCs. ATM phosphorylation and γ‐H2AX and phospho‐p53 increased in early‐passage MSCs while decreased in late‐passage MSCs. Through inhibition by KU55933, DDR pathway in early‐passage MSCs was shown to be ATM‐dependent. Higher levels of poly (ADP‐ribose) polymerase‐1 (PARP‐1) and PAR synthesis were observed in early‐passage MSCs than in late‐passage MSCs. Knockdown of PARP‐1 in early‐passage MSCs resulted in sensitization to irradiation‐induced apoptosis. Overexpression of PARP‐1 in late passage MSCs could render irradiation resistance. Lower activity of DDR in late‐passage MSCs was associated with rapid proteasomal degradation of PARP‐1. In conclusion, early‐passage MSCs are more irradiation‐resistant and have increased DDR activity involving PARP‐1, ATM and their downstream signals. Stem Cells Translational Medicine2017;6:1504–1514
Collapse
Affiliation(s)
- Po-Kuei Wu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Orthopaedics & Traumatology, Taipei Veterans General Hospital, Taiwan.,Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taiwan
| | - Jir-You Wang
- Department of Orthopaedics & Traumatology, Taipei Veterans General Hospital, Taiwan.,Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taiwan.,Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Fong Chen
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Orthopaedics & Traumatology, Taipei Veterans General Hospital, Taiwan.,Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taiwan
| | - Kuang-Yu Chao
- Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taiwan
| | - Ming-Chau Chang
- Department of Orthopaedics & Traumatology, Taipei Veterans General Hospital, Taiwan
| | - Wei-Ming Chen
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Orthopaedics & Traumatology, Taipei Veterans General Hospital, Taiwan.,Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taiwan
| | - Shih-Chieh Hung
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Orthopaedics & Traumatology, Taipei Veterans General Hospital, Taiwan.,Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Integrative Stem Cell Center, Chinese Medical University Hospital, Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
20
|
Li J, Liu W, Yao W. Immortalized Human Bone Marrow Derived Stromal Cells in Treatment of Transient Cerebral Ischemia in Rats. J Alzheimers Dis 2019; 69:871-880. [PMID: 31156178 DOI: 10.3233/jad-190279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jianyuan Li
- Neurosurgical Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Neurosurgical Department, Rizhao City Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Weidong Liu
- Neurosurgical Department, Liaocheng People’s Hospital, Liaocheng, China
| | - Weicheng Yao
- Neurosurgical Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
21
|
Abstract
Mesenchymal stem cells (MSCs) are multipotent tissue stem cells that differentiate into a number of mesodermal tissue types, including osteoblasts, adipocytes, chondrocytes and myofibroblasts. MSCs were originally identified in the bone marrow (BM) of humans and other mammals, but recent studies have shown that they are multilineage progenitors in various adult organs and tissues. MSCs that localize at perivascular sites function to rapidly respond to external stimuli and coordinate with the vascular and immune systems to accomplish the wound healing process. Cancer, considered as wounds that never heal, is also accompanied by changes in MSCs that parallels the wound healing response. MSCs are now recognized as key players at distinct steps of tumorigenesis. In this review, we provide an overview of the function of MSCs in wound healing and cancer progression with the goal of providing insight into the development of novel MSC-manipulating strategies for clinical cancer treatment.
Collapse
|
22
|
Bajelan B, Zaki‐Dizaji M, Darabi S, Rajaei F. The effects of Nutlin‐3 on morphology, cellular proliferation, and apoptosis in rat primary mesenchymal stem cells. J Cell Physiol 2018; 234:11424-11430. [DOI: 10.1002/jcp.27798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Babak Bajelan
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences Qazvin Iran
| | - Majid Zaki‐Dizaji
- Department of Medical Genetics School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences Qazvin Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences Qazvin Iran
- Department of Histology and Embryology, Faculty of Medicine Qazvin University of Medical Sciences Qazvin Iran
| |
Collapse
|
23
|
Rühle A, Huber PE, Saffrich R, Lopez Perez R, Nicolay NH. The current understanding of mesenchymal stem cells as potential attenuators of chemotherapy-induced toxicity. Int J Cancer 2018; 143:2628-2639. [PMID: 29931767 DOI: 10.1002/ijc.31619] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022]
Abstract
Chemotherapeutic agents are part of the standard treatment algorithms for many malignancies; however, their application and dosage are limited by their toxic effects to normal tissues. Chemotherapy-induced toxicities can be long-lasting and may be incompletely reversible; therefore, causative therapies for chemotherapy-dependent side effects are needed, especially considering the increasing survival rates of treated cancer patients. Mesenchymal stem cells (MSCs) have been shown to exhibit regenerative abilities for various forms of tissue damage. Preclinical data suggest that MSCs may also help to alleviate tissue lesions caused by chemotherapeutic agents, mainly by establishing a protective microenvironment for functional cells. Due to the systemic administration of most anticancer agents, the effects of these drugs on the MSCs themselves are of crucial importance to use stem cell-based approaches for the treatment of chemotherapy-induced tissue toxicities. Here, we present a concise review of the published data regarding the influence of various classes of chemotherapeutic agents on the survival, stem cell characteristics and physiological functions of MSCs. Molecular mechanisms underlying the effects are outlined, and resulting challenges of MSC-based treatments for chemotherapy-induced tissue injuries are discussed.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Rainer Saffrich
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany.,Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
24
|
Baxter-Holland M, Dass CR. Doxorubicin, mesenchymal stem cell toxicity and antitumour activity: implications for clinical use. ACTA ACUST UNITED AC 2018; 70:320-327. [PMID: 29355940 DOI: 10.1111/jphp.12869] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/25/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The use of doxorubicin, an antineoplastic medication used for the treatment of cancers via mechanisms that prevent replication of cells or lead to their death, can result in damage to healthy cells as well as malignant. Among the affected cells are mesenchymal stem cells (MSCs), which are involved in the maintenance and repair of tissues in the body. This review explores the mechanisms of biological effects and damage attributed to doxorubicin on MSCs. The PubMed database was used as a source of literature for this review. KEY FINDINGS Doxorubicin has the potential to lead to significant and irreversible damage to the human bone marrow environment, including MSCs. The primary known mechanism of these changes is through free radical damage and activation of apoptotic pathways. The presence of MSCs in culture or in vivo appears to either suppress or promote tumour growth. Interactions between doxorubicin and MSCs have the potential to increase chemotherapy resistance. SUMMARY Doxorubicin-induced damage to MSCs is of concern clinically. However, MSCs also have been associated with resistance of tumour cells to drugs including doxorubicin. Further studies, particularly in vivo, are needed to provide consistent results of how the doxorubicin-induced changes to MSCs affect treatment and patient health.
Collapse
Affiliation(s)
- Mia Baxter-Holland
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia.,Curtin Health Innovation Research Institute, Perth, WA, Australia
| |
Collapse
|
25
|
Münz F, Lopez Perez R, Trinh T, Sisombath S, Weber KJ, Wuchter P, Debus J, Saffrich R, Huber PE, Nicolay NH. Human mesenchymal stem cells lose their functional properties after paclitaxel treatment. Sci Rep 2018; 8:312. [PMID: 29321693 PMCID: PMC5762916 DOI: 10.1038/s41598-017-18862-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an integral part of the bone marrow niche and aid in the protection, regeneration and proliferation of hematopoietic stem cells after exposure to myelotoxic taxane anti-cancer agents, but the influence of taxane compounds on MSCs themselves remains incompletely understood. Here, we show that bone marrow-derived MSCs are highly sensitive even to low concentrations of the prototypical taxane compound paclitaxel. While MSCs remained metabolically viable, they were strongly impaired regarding both their proliferation and their functional capabilities after exposure to paclitaxel. Paclitaxel treatment resulted in reduced cell migration, delays in cellular adhesion and significant dose-dependent inhibition of the stem cells’ characteristic multi-lineage differentiation potential. Cellular morphology and expression of the defining surface markers remained largely unaltered. Paclitaxel only marginally increased apoptosis in MSCs, but strongly induced premature senescence in these stem cells, thereby explaining the preservation of the metabolic activity of functionally inactivated MSCs. The reported sensitivity of MSC function to paclitaxel treatment may help to explain the severe bone marrow toxicities commonly caused by taxane-based anti-cancer treatments.
Collapse
Affiliation(s)
- Franziska Münz
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ramon Lopez Perez
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Thuy Trinh
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Klaus-Josef Weber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Friedrich-Ebert-Str. 107, 68167, Mannheim, Germany
| | - Jürgen Debus
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Friedrich-Ebert-Str. 107, 68167, Mannheim, Germany.,Department of Hematology and Oncology, Heidelberg University Hospital, Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter E Huber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Nils H Nicolay
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| |
Collapse
|
26
|
Rühle A, Perez RL, Glowa C, Weber KJ, Ho AD, Debus J, Saffrich R, Huber PE, Nicolay NH. Cisplatin radiosensitizes radioresistant human mesenchymal stem cells. Oncotarget 2017; 8:87809-87820. [PMID: 29152122 PMCID: PMC5675674 DOI: 10.18632/oncotarget.21214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022] Open
Abstract
Cisplatin-based chemo-radiotherapy is widely used to treat cancers with often severe therapy-associated late toxicities. While mesenchymal stem cells (MSCs) were shown to aid regeneration of cisplatin- or radiation-induced tissue lesions, the effect of the combined treatment on the stem cells remains unknown. Here we demonstrate that cisplatin treatment radiosensitized human bone marrow-derived MSCs in a dose-dependent manner and increased levels of radiation-induced apoptosis. However, the defining stem cell properties of MSCs remained largely intact after cisplatin-based chemo-radiation, and stem cell motility, adhesion, surface marker expression and the characteristic differentiation potential were not significantly influenced. The increased cisplatin-mediated radiosensitivity was associated with a cell cycle shift of MSCs towards the radiosensitive G2/M phase and increased residual DNA double-strand breaks. These data demonstrate for the first time a dose-dependent radiosensitization effect of MSCs by cisplatin. Clinically, the observed increase in radiation sensitivity and subsequent loss of regenerative MSCs may contribute to the often severe late toxicities observed after cisplatin-based chemo-radiotherapy in cancer patients.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany
| | - Christin Glowa
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany
| | - Klaus-Josef Weber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Department of Hematology and Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Peter E Huber
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
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: 0.9] [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
|
28
|
Resistance for Genotoxic Damage in Mesenchymal Stromal Cells Is Increased by Hypoxia but Not Generally Dependent on p53-Regulated Cell Cycle Arrest. PLoS One 2017; 12:e0169921. [PMID: 28081228 PMCID: PMC5231334 DOI: 10.1371/journal.pone.0169921] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/23/2016] [Indexed: 11/19/2022] Open
Abstract
Adult stem cells including multipotent mesenchymal stromal cells (MSC) acquire a high amount of DNA-damage due to their prolonged lifespan. MSC may exert specific mechanisms of resistance to avoid loss of functional activity. We have previously shown that resistance of MSC is associated with an induction of p53 and proliferation arrest upon genotoxic damage. Hypoxia may also contribute to resistance in MSC due to the low oxygen tension in the niche. In this study we characterized the role of p53 and contribution of hypoxia in resistance of MSC to genotoxic damage. MSC exhibited increased resistance to cisplatin induced DNA-damage. This resistance was associated with a temporary G2/M cell cycle arrest, induction of p53- and p21-expression and reduced cyclin B / cdk1-levels upon subapoptotic damage. Resistance of MSC to cisplatin was increased at hypoxic conditions i. e. oxygen <0.5%. However, upon hypoxia the cisplatin-induced cell cycle arrest and expression of p53 and p21 were abrogated. MSC with shRNA-mediated p53 knock-down showed a reduced cell cycle arrest and increased cyclin B / cdk1 expression. However, this functional p53 knock down did not alter the resistance to cisplatin. In contrast to cisplatin, functional p53-knock-down increased the resistance of MSC to etoposide. We conclude that resistance of MSC to genotoxic damage is influenced by oxygen tension but is not generally dependent on p53. Thus, p53-dependent and p53-independent mechanisms of resistance are likely to contribute to the life-long functional activity of MSC in vivo. These findings indicate that hypoxia and different resistance pathways contribute to the phenotype that enables the prolonged lifespan of MSC.
Collapse
|
29
|
Harris WM, Zhang P, Plastini M, Ortiz T, Kappy N, Benites J, Alexeev E, Chang S, Brockunier R, Carpenter JP, Brown SA. Evaluation of function and recovery of adipose-derived stem cells after exposure to paclitaxel. Cytotherapy 2016; 19:211-221. [PMID: 27887867 DOI: 10.1016/j.jcyt.2016.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/28/2016] [Accepted: 10/19/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS Adipose-derived stem cells (ASCs) are considered to play a positive role in wound healing as evidenced by their increasing use in breast reconstructive procedures. After chemotherapy for breast cancer, poor soft tissue wound healing is a major problem. In the present study, the functional capabilities and recovery of ASCs after exposure to chemotherapeutic agent paclitaxel (PTX) using in vitro and ex vivo models were demonstrated. METHODS Human ASCs were isolated from periumbilical fat tissue and treated with PTX at various concentrations. Adult Sprague-Dawley rats were given intravenous injections with PTX. Two and four weeks after the initial PTX treatment, ASCs were isolated from rat adipose tissue. Proliferation, cell viability, apoptosis and cell migration rates were measured by growth curves, MTT assays, flow cytometry and scratch assays. ASCs were cultured in derivative-specific differentiation media with or without PTX for 3 weeks. Adipogenic, osteogenic and endothelial differentiation levels were measured by quantitative reverse transcriptase polymerase chain reaction and histological staining. RESULTS PTX induced apoptosis, decreased the proliferation and cell migration rates of ASCs and inhibited ASCs multipotent differentiation in both in vitro human ASC populations and ex vivo rat ASC populations with PTX treatment. Furthermore, after cessation of PTX, ASCs exhibited recovery potential of differentiation capacity in both in vitro and animal studies. CONCLUSIONS Our results provide insight into poor soft tissue wound healing and promote further understanding of the potential capability of ASCs to serve as a cell source for fat grafting and reconstruction in cancer patients undergoing chemotherapy treatment.
Collapse
Affiliation(s)
- William M Harris
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Ping Zhang
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA.
| | - Michael Plastini
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Telisha Ortiz
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Nikolas Kappy
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Jefferson Benites
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Edward Alexeev
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Shaohua Chang
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Ross Brockunier
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | | | - Spencer A Brown
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| |
Collapse
|
30
|
Mesenchymal stem cells are sensitive to bleomycin treatment. Sci Rep 2016; 6:26645. [PMID: 27215195 PMCID: PMC4877675 DOI: 10.1038/srep26645] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/06/2016] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to attenuate pulmonary damage induced by bleomycin-based anticancer treatments, but the influence of bleomycin on the stem cells themselves remains largely unknown. Here, we demonstrate that human bone marrow-derived MSCs are relatively sensitive to bleomycin exposure compared to adult fibroblasts. MSCs revealed increased levels of apoptosis after bleomycin treatment, while cellular morphology, stem cell surface marker expression and the ability for adhesion and migration remained unchanged. Bleomycin treatment also resulted in a reduced adipogenic differentiation potential of these stem cells. MSCs were found to efficiently repair DNA double strand breaks induced by bleomycin, mostly through non-homologous end joining repair. Low mRNA and protein expression levels of the inactivating enzyme bleomycin hydrolase were detected in MSCs that may contribute to the observed bleomycin-sensitive phenotype of these cells. The sensitivity of MSCs against bleomycin needs to be taken into consideration for ongoing and future treatment protocols investigating these stem cells as a potential treatment option for bleomycin-induced pulmonary damage in the clinic.
Collapse
|
31
|
Bellagamba BC, Abreu BRRD, Grivicich I, Markarian CF, Chem E, Camassola M, Nardi NB, Dihl RR. Human mesenchymal stem cells are resistant to cytotoxic and genotoxic effects of cisplatin in vitro. Genet Mol Biol 2016; 39:129-34. [PMID: 27007906 PMCID: PMC4807379 DOI: 10.1590/1678-4685-gmb-2015-0057] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 08/11/2015] [Indexed: 11/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known for their important properties involving multilineage differentiation potential., trophic factor secretion and localization along various organs and tissues. On the dark side, MSCs play a distinguished role in tumor microenvironments by differentiating into tumor-associated fibroblasts or supporting tumor growth via distinct mechanisms. Cisplatin (CIS) is a drug widely applied in the treatment of a large number of cancers and is known for its cytotoxic and genotoxic effects, both in vitro and in vivo. Here we assessed the effects of CIS on MSCs and the ovarian cancer cell line OVCAR-3, by MTT and comet assays. Our results demonstrated the resistance of MSCs to cell death and DNA damage induction by CIS, which was not observed when OVCAR-3 cells were exposed to this drug.
Collapse
Affiliation(s)
| | | | | | | | - Eduardo Chem
- Complexo Hospitalar Santa Casa de Porto Alegre, Porto Alegre, RS, Brazil
| | | | | | | |
Collapse
|
32
|
Luetzkendorf J, Nerger K, Hering J, Moegel A, Hoffmann K, Hoefers C, Mueller-Tidow C, Mueller LP. Cryopreservation does not alter main characteristics of Good Manufacturing Process-grade human multipotent mesenchymal stromal cells including immunomodulating potential and lack of malignant transformation. Cytotherapy 2016; 17:186-98. [PMID: 25593077 DOI: 10.1016/j.jcyt.2014.10.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/07/2014] [Accepted: 10/21/2014] [Indexed: 01/16/2023]
Abstract
BACKGROUND AIMS The immunomodulating capacity of multipotent mesenchymal stromal cells (MSCs) qualifies them as a therapeutic tool in several diseases. However, repeated transplantation with products of reproducible characteristics may be required. This could be achieved with cryopreserved aliquots of Good Manufacturing Practice (GMP)-grade MSCs. However, the impact of cryopreservation on the characteristics of GMP-MSCs is ill defined. METHODS We produced fresh and cryopreserved MSCs from human donors with a xenogen-free GMP protocol. Immunogenicity and immunomodulating capacity were tested in co-culture with putative recipient-specific peripheral blood mononuclear cells (PBMCs). Risk of malignant transformation was assessed in vitro and in vivo. RESULTS Cryopreservation had no impact on viability and consensus criteria of MSCs. In co-culture with PBMCs, MSCs showed low immunogenicity and suppressed mitogen-stimulated proliferation of PBMC irrespective of cryopreservation. Cytogenetic aberrations were not observed consistently in fresh and cryopreserved products, and no signs of malignant transformation occurred in functional assays. MSC products from an elderly pretreated donor showed reduced functional quality, but imminent failure of functional criteria could be detected by an increased population doubling time in early passages. DISCUSSION This study is the first systematic analysis on cryopreservation of xenogen-free human bone marrow-derived GMP-MSCs. The data support that cryopreservation does not alter the characteristics of the cells and thus may allow the generation of products for serial transplantation. In addition, the protocol allowed early detection of MSC products with low functional capacity.
Collapse
Affiliation(s)
- Jana Luetzkendorf
- Universitätsklinik und Poliklinik für Innere Medizin IV, Halle (Saale), Germany
| | - Katrin Nerger
- Universitätsklinik und Poliklinik für Innere Medizin IV, Halle (Saale), Germany
| | - Julian Hering
- Einrichtung für Transfusionsmedizin, Halle (Saale), Germany
| | | | - Katrin Hoffmann
- Institut für Humangenetik, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | - Christiane Hoefers
- Institut für Humangenetik, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | | | - Lutz P Mueller
- Universitätsklinik und Poliklinik für Innere Medizin IV, Halle (Saale), Germany.
| |
Collapse
|
33
|
Nicolay NH, Rühle A, Perez RL, Trinh T, Sisombath S, Weber KJ, Schmezer P, Ho AD, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells exhibit resistance to topoisomerase inhibition. Cancer Lett 2016; 374:75-84. [PMID: 26876302 DOI: 10.1016/j.canlet.2016.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Inhibition of cellular topoisomerases has been established as an effective way of treating certain cancers, albeit with often high levels of toxicity to the bone marrow. While the involvement of mesenchymal stem cells (MSCs) in bone marrow homeostasis and regeneration has been well established, the effects of topoisomerase-inhibiting anticancer agents remain largely unknown. MATERIALS AND METHODS Human bone marrow MSCs were treated with topoisomerase I inhibitor irinotecan or topoisomerase II inhibitor etoposide, and survival and apoptosis levels were measured. The influence of topoisomerase inhibition on cellular morphology, adhesion and migration potential and the ability to differentiate was assessed. Additionally, the role of individual DNA double-strand break repair pathways in MSCs was investigated as a potential cellular mechanism of resistance to topoisomerase inhibitors. RESULTS Human bone marrow MSCs were found relatively resistant to topoisomerase I and II inhibitors and show survival levels comparable to these of differentiated fibroblasts. Treatment with irinotecan or etoposide did not significantly influence cellular adhesion, migratory ability, surface marker expression or induction of apoptosis in human MSCs. The ability to differentiate was found preserved in MSCs after exposure to high doses of irinotecan or etoposide. MSCs were able to efficiently repair DNA double-strand breaks induced by topoisomerase inhibitors both by non-homologous end joining and homologous recombination pathways. CONCLUSION Our data demonstrate a topoisomerase-resistant phenotype of human MSCs that may at least in part be due to the stem cells' ability to efficiently remove DNA damage caused by these anticancer agents. The observed resistance of MSCs warrants further investigation of these cells as a potential therapeutic option for treating topoisomerase inhibitor-induced bone marrow damage.
Collapse
Affiliation(s)
- Nils H Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Alexander Rühle
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Peter Schmezer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Mesenchymal stem cells maintain their defining stem cell characteristics after treatment with cisplatin. Sci Rep 2016; 6:20035. [PMID: 26805490 PMCID: PMC4726328 DOI: 10.1038/srep20035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 12/23/2015] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) aid the regeneration of tissues damaged by treatment with cisplatin. However, the effects of this cytotoxic drug on the stem cells have been largely unknown. Here we demonstrate that human bone marrow-derived MSCs are relatively resistant to cisplatin treatment and show resistance levels comparable to these of differentiated fibroblasts. Cisplatin did not affect cellular morphology, adhesion or induction of apoptosis in MSCs. The potential for differentiation was preserved after exposure to cisplatin, and established MSC surface markers were observed to be stably expressed irrespective of cisplatin treatment. Cytoskeletal rearrangements and high expression levels of individual heat shock proteins were detected in MSCs and may be partly responsible for the observed cisplatin resistance. The cisplatin-resistant phenotype of human MSCs supports the concept of further investigating these stem cells as a potential treatment option for cisplatin-induced tissue damage.
Collapse
|
35
|
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: 72] [Impact Index Per Article: 8.0] [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
|
36
|
Widder M, Lützkendorf J, Caysa H, Unverzagt S, Wickenhauser C, Benndorf RA, Schmoll HJ, Müller-Tidow C, Müller T, Müller LP. Multipotent mesenchymal stromal cells promote tumor growth in distinct colorectal cancer cells by a β1-integrin-dependent mechanism. Int J Cancer 2015; 138:964-75. [PMID: 26356035 DOI: 10.1002/ijc.29844] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 12/26/2022]
Abstract
Tumor-stroma interactions play an essential role in the biology of colorectal carcinoma (CRC). Multipotent mesenchymal stromal cells (MSC) may represent a pivotal part of the stroma in CRC, but little is known about the specific interaction of MSC with CRC cells derived from tumors with different mutational background. In previous studies we observed that MSC promote the xenograft growth of the CRC cell-line DLD1. In the present study, we aimed to analyze the mechanisms of MSC-promoted tumor growth using various in vitro and in vivo experimental models and CRC cells of different mutational status. MSC specifically interacted with distinct CRC cells and supported tumor seeding in xenografts. The MSC-CRC interaction facilitated three-dimensional spheroid formation in CRC cells with dysfunctional E-cadherin system. Stable knock-downs revealed that the MSC-facilitated spheroid formation depended on β1-integrin in CRC cells. Specifically in α-catenin-deficient CRC cells this β1-integrin-dependent interaction resulted in a MSC-mediated promotion of early tumor growth in vivo. Collagen I and other extracellular matrix compounds were pivotal for the functional MSC-CRC interaction. In conclusion, our data demonstrate a differential interaction of MSC with CRC cells of different mutational background. Our study is the first to show that MSC specifically compared to normal fibroblasts impact early xenograft growth of distinct α-catenin deficient CRC cells possibly through secretion of extracellular matrix. This mechanism could serve as a future target for therapy and metastasis prevention.
Collapse
Affiliation(s)
- Miriam Widder
- Universitätsklinik und Poliklinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Jana Lützkendorf
- Universitätsklinik und Poliklinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Henrike Caysa
- Institut für Pharmazie, Institutsbereich Pharmazeutische Technologie und Biopharmazie, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Susanne Unverzagt
- Institut für Medizinische Epidemiologie, Biometrie und Informatik, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Claudia Wickenhauser
- Institut für Pathologie, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Ralf A Benndorf
- Institut für Pharmazie, Institutsbereich Pharmazeutische Chemie und Klinische Pharmazie, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Hans-Joachim Schmoll
- Universitätsklinik und Poliklinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Carsten Müller-Tidow
- Universitätsklinik und Poliklinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Thomas Müller
- Universitätsklinik und Poliklinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Lutz P Müller
- Universitätsklinik und Poliklinik für Innere Medizin IV, Hämatologie und Onkologie, Universitätsklinikum Halle, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| |
Collapse
|
37
|
Magnucki G, Schenk U, Ahrens S, Navarrete Santos A, Gernhardt CR, Schaller HG, Hoang-Vu C. Expression of the IGF-1, IGFBP-3 and IGF-1 receptors in dental pulp stem cells and impacted third molars. J Oral Sci 2015; 55:319-27. [PMID: 24351920 DOI: 10.2334/josnusd.55.319] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
IGF-1 regulates the metabolism of hard dental tissue through binding to the IGF-1 receptor on target cells. Furthermore, IGF-binding-protein-3 promotes the accessibility of IGF-1. The aim of this study was to investigate the expression of IGF-1, IGFBP-3 and IGF-1R in STRO-1-positive dental pulp stem cells (DPSCs) and fully impacted wisdom teeth in relation to tooth development. Third molars were surgically removed from 60 patients and classified into two groups: teeth showing ongoing development (group 1) and teeth that had completed root shaping (group 2). The transcript and protein levels of IGF-1, IGFBP-3 and IGF-1R were investigated using RT-PCR and immunohistochemistry. The expression of the same proteins was also analyzed in DPSCs. The teeth from group 1 showed significantly stronger expression of IGF-1 and IGF-1R. The major sources of all of the proteins investigated immunohistochemically in sections of wisdom teeth were odontoblasts, cementoblasts and cell colonies in the pulpal mesenchyme. These colonies were identified as stem cells in view of their positivity for STRO-1, and the cells were subsequently sorted by flow cytometry. These DPSCs demonstrated high levels of pluripotency markers and IGF-1 and IGF-1R. We conclude that members of the IGF-1 family are involved in the late stage of tooth development and the process of pulpal differentiation.
Collapse
Affiliation(s)
- Gabriel Magnucki
- Department of Operative Dentistry and Periodontology, University School of Dental Medicine, Faculty of Medicine, Martin-Luther-University of Halle-Wittenberg
| | | | | | | | | | | | | |
Collapse
|
38
|
Krüger M, Krüger JP, Kinne RW, Kaps C, Endres M. Are surface antigens suited to verify the redifferentiation potential and culture purity of human chondrocytes in cell-based implants. Tissue Cell 2015; 47:489-97. [PMID: 26254705 DOI: 10.1016/j.tice.2015.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/15/2015] [Accepted: 07/15/2015] [Indexed: 11/18/2022]
Abstract
Cell expansion in vitro is a prequisite to obtain a sufficient quantity of cells for cell-based cartilage repair of articular cartilage lesions. During this process verification of redifferentiation potential of highly expanded chondrocytes is required. Furthermore, cellular impurities of chondrocyte cultures have to be excluded. For this purpose, redifferentiation of expanded human chondrocytes in passage 3 or 5 was initiated in bioresorbable polyglycolic acid-fibrin (PGA-fibrin) scaffolds and selected potential markers were analysed during the process of cell expansion and redifferentiation. Chondrocyte expansion was accompanied by a decrease of collagen type II and COMP and an increase of collagen type I expression indicating cell dedifferentiation. Redifferentiation of chondrocytes in PGA-fibrin scaffolds was accompanied by an increase of collagen II/I ratio. Flow cytometric analyses revealed that in contrast to CD44 and CD49e, CD63 and CD166 showed significant changes in the number of positive cells during redifferentiation. CD14 and CD45 are not expressed by chondrocytes and are therefore possible candidates to detect specifically monocytes or haematopoetic cells in chondrocyte cultures. Characterization of surface antigen expression revealed two promising candidates (CD63 and CD166) to describe the process of redifferentiation, while CD14 and CD45 are suitable markers to exclude impurities by monocytes or haematopoetic cells.
Collapse
Affiliation(s)
- M Krüger
- TransTissue Technologies GmbH, Berlin, Germany
| | - J P Krüger
- TransTissue Technologies GmbH, Berlin, Germany; Department of Rheumatology, Laboratory for Tissue Engineering, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - R W Kinne
- Department of Orthopedics, Experimental Rheumatology Unit, Friedrich Schiller University, Jena, Germany
| | - C Kaps
- TransTissue Technologies GmbH, Berlin, Germany
| | - M Endres
- TransTissue Technologies GmbH, Berlin, Germany; Department of Rheumatology, Laboratory for Tissue Engineering, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
39
|
Bosco DB, Kenworthy R, Zorio DAR, Sang QXA. Human mesenchymal stem cells are resistant to Paclitaxel by adopting a non-proliferative fibroblastic state. PLoS One 2015; 10:e0128511. [PMID: 26029917 PMCID: PMC4452335 DOI: 10.1371/journal.pone.0128511] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/29/2015] [Indexed: 12/31/2022] Open
Abstract
Human mesenchymal stem cell (hMSC) resistance to the apoptotic effects of chemotherapeutic drugs has been of major interest, as these cells can confer this resistance to tumor microenvironments. However, the effects of internalized chemotherapeutics upon hMSCs remain largely unexplored. In this study, cellular viability and proliferation assays, combined with different biochemical approaches, were used to investigate the effects of Paclitaxel exposure upon hMSCs. Our results indicate that hMSCs are highly resistant to the cytotoxic effects of Paclitaxel treatment, even though there was no detectable expression of the efflux pump P-glycoprotein, the usual means by which a cell resists Paclitaxel treatment. Moreover, Paclitaxel treatment induces hMSCs to adopt a non-proliferative fibroblastic state, as evidenced by changes to morphology, cellular markers, and a reduction in differentiation potential that is not directly coupled to the cytoskeletal effects of Paclitaxel. Taken together, our results show that Paclitaxel treatment does not induce apoptosis in hMSCs, but does induce quiescence and phenotypic changes.
Collapse
Affiliation(s)
- Dale B. Bosco
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States of America
| | - Rachael Kenworthy
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States of America
| | - Diego A. R. Zorio
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States of America
- * E-mail: (DARZ); (QXAS)
| | - Qing-Xiang Amy Sang
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, United States of America
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, United States of America
- * E-mail: (DARZ); (QXAS)
| |
Collapse
|
40
|
Minieri V, Saviozzi S, Gambarotta G, Lo Iacono M, Accomasso L, Cibrario Rocchietti E, Gallina C, Turinetto V, Giachino C. Persistent DNA damage-induced premature senescence alters the functional features of human bone marrow mesenchymal stem cells. J Cell Mol Med 2015; 19:734-43. [PMID: 25619736 PMCID: PMC4395188 DOI: 10.1111/jcmm.12387] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/24/2014] [Indexed: 12/26/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are adult multipotent stem cells located in various tissues, including the bone marrow. In contrast to terminally differentiated somatic cells, adult stem cells must persist and function throughout life to ensure tissue homeostasis and repair. For this reason, they must be equipped with DNA damage responses able to maintain genomic integrity while ensuring their lifelong persistence. Evaluation of hMSC response to genotoxic insults is of great interest considering both their therapeutic potential and their physiological functions. This study aimed to investigate the response of human bone marrow MSCs to the genotoxic agent Actinomycin D (ActD), a well-known anti-tumour drug. We report that hMSCs react by undergoing premature senescence driven by a persistent DNA damage response activation, as hallmarked by inhibition of DNA synthesis, p21 and p16 protein expression, marked Senescent Associated β-galactosidase activity and enlarged γH2AX foci co-localizing with 53BP1 protein. Senescent hMSCs overexpress several senescence-associated secretory phenotype (SASP) genes and promote motility of lung tumour and osteosarcoma cell lines in vitro. Our findings disclose a multifaceted consequence of ActD treatment on hMSCs that on the one hand helps to preserve this stem cell pool and prevents damaged cells from undergoing neoplastic transformation, and on the other hand alters their functional effects on the surrounding tissue microenvironment in a way that might worsen their tumour-promoting behaviour.
Collapse
Affiliation(s)
- Valentina Minieri
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Stem cells: the pursuit of genomic stability. Int J Mol Sci 2014; 15:20948-67. [PMID: 25405730 PMCID: PMC4264205 DOI: 10.3390/ijms151120948] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/02/2014] [Accepted: 11/04/2014] [Indexed: 12/18/2022] Open
Abstract
Stem cells harbor significant potential for regenerative medicine as well as basic and clinical translational research. Prior to harnessing their reparative nature for degenerative diseases, concerns regarding their genetic integrity and mutation acquisition need to be addressed. Here we review pluripotent and multipotent stem cell response to DNA damage including differences in DNA repair kinetics, specific repair pathways (homologous recombination vs. non-homologous end joining), and apoptotic sensitivity. We also describe DNA damage and repair strategies during reprogramming and discuss potential genotoxic agents that can reduce the inherent risk for teratoma formation and mutation accumulation. Ensuring genomic stability in stem cell lines is required to achieve the quality control standards for safe clinical application.
Collapse
|
42
|
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: 1.6] [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
|
43
|
Buravkova LB, Andreeva ER, Gogvadze V, Zhivotovsky B. Mesenchymal stem cells and hypoxia: where are we? Mitochondrion 2014; 19 Pt A:105-12. [PMID: 25034305 DOI: 10.1016/j.mito.2014.07.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/09/2014] [Indexed: 12/23/2022]
Abstract
Multipotent mesenchymal stromal cells (MSCs) are involved in the organization and maintenance of tissue integrity. MSCs have also attracted attention as a promising tool for cell therapy and regenerative medicine. However, their usage is limited due to cell impairment induced by an extremely harsh microenvironment during transplantation ex vivo. The microenvironment of MSCs in tissue depots is characterized by rather low oxygen consumption, demonstrating that MSCs might be quite resistant to oxygen limitation. However, accumulated data revealed that the response of MSCs to hypoxic conditions is rather controversial, demonstrating both damaging and ameliorating effects. Here, we make an attempt to summarize recent knowledge on the survival of MSCs under low oxygen conditions of varying duration and severity and to elucidate the mechanisms of MSC resistance/sensitivity to hypoxic impact.
Collapse
Affiliation(s)
- L B Buravkova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia; Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia
| | - E R Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - V Gogvadze
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden
| | - B Zhivotovsky
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden.
| |
Collapse
|
44
|
Wuchter P, Bieback K, Schrezenmeier H, Bornhäuser M, Müller LP, Bönig H, Wagner W, Meisel R, Pavel P, Tonn T, Lang P, Müller I, Renner M, Malcherek G, Saffrich R, Buss EC, Horn P, Rojewski M, Schmitt A, Ho AD, Sanzenbacher R, Schmitt M. Standardization of Good Manufacturing Practice-compliant production of bone marrow-derived human mesenchymal stromal cells for immunotherapeutic applications. Cytotherapy 2014; 17:128-39. [PMID: 24856898 DOI: 10.1016/j.jcyt.2014.04.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 03/26/2014] [Accepted: 04/05/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND AIMS Human mesenchymal stem or stromal cells (MSCs) represent a potential resource not only for regenerative medicine but also for immunomodulatory cell therapies. The application of different MSC culture protocols has significantly hampered the comparability of experimental and clinical data from different laboratories and has posed a major obstacle for multicenter clinical trials. Manufacturing of cell products for clinical application in the European Community must be conducted in compliance with Good Manufacturing Practice and requires a manufacturing license. In Germany, the Paul-Ehrlich-Institut as the Federal Authority for Vaccines and Biomedicines is critically involved in the approval process. METHODS This report summarizes a consensus meeting between researchers, clinicians and regulatory experts on standard quality requirements for MSC production. RESULTS The strategy for quality control testing depends on the product's cell composition, the manufacturing process and the indication and target patient population. Important quality criteria in this sense are, among others, the immunophenotype of the cells, composition of the culture medium and the risk for malignant transformation, as well as aging and the immunosuppressive potential of the manufactured MSCs. CONCLUSIONS This position paper intends to provide relevant information to interested parties regarding these criteria to foster the development of scientifically valid and harmonized quality standards and to support approval of MSC-based investigational medicinal products.
Collapse
Affiliation(s)
- Patrick Wuchter
- Department of Medicine V, Heidelberg University, Heidelberg, Germany.
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology Mannheim, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University of Ulm, Ulm, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Lutz P Müller
- Department of Medicine IV, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Halvard Bönig
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt/Main and German Red Cross Blood Service Baden-Württemberg-Hessen, Frankfurt/Main, Germany
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Technology, Stem Cell Biology and Cellular Engineering, University of Aachen Medical School, Aachen, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Clinic for Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Petra Pavel
- Stem Cell Laboratory, IKTZ Heidelberg GmbH, Heidelberg, Germany
| | - Torsten Tonn
- Institute of Transfusion Medicine, Red Cross Blood Transfusion Service Dresden, Dresden, Germany
| | - Peter Lang
- Department of Pediatrics, University Clinic Tübingen, Tübingen, Germany
| | - Ingo Müller
- Clinic for Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Renner
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Georg Malcherek
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Rainer Saffrich
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Eike C Buss
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Patrick Horn
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Markus Rojewski
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University of Ulm, Ulm, Germany
| | - Anita Schmitt
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Anthony D Ho
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Ralf Sanzenbacher
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Michael Schmitt
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
45
|
Oliver L, Hue E, Séry Q, Lafargue A, Pecqueur C, Paris F, Vallette FM. Differentiation-related response to DNA breaks in human mesenchymal stem cells. Stem Cells 2013; 31:800-7. [PMID: 23341263 DOI: 10.1002/stem.1336] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 11/25/2012] [Indexed: 12/19/2022]
Abstract
We have recently shown that the in vitro differentiation of human mesenchymal stem cells (hMSCs) was accompanied by an increased sensitivity toward apoptosis; however, the mechanism responsible for this shift is not known. Here, we show that the repair of DNA double-strand breaks (DSBs) was more rapid in undifferentiated hMSCs than in differentiated osteoblasts by quantification of the disappearance of γ-H2AX foci in the nuclei after γ-irradiation-induced DNA damage. In addition, there was a marked and prolonged increase in the level of nuclear Ku70 and an increased phosphorylation of DNA-PKcs. This was accompanied by an augmentation in the phosphorylation of ATM in hMSCs post-irradiation suggesting the nonhomologous end joining repair mechanism. However, when hMSCs were induced to differentiate along the osteogenic or adipogenic pathways; irradiation of these cells caused an expeditious and robust cell death, which was primarily apoptotic. This was in sharp contrast to undifferentiated hMSCs, which were highly resistant to irradiation and/or temozolomide-induced DSBs. In addition, we observed a 95% recovery from DSB in these cells. Our results suggest that apoptosis and DNA repair are major safeguard mechanisms in the control of hMSCs differentiation after DNA damage.
Collapse
Affiliation(s)
- Lisa Oliver
- CRCNA-INSERM UMR 892-CNRS UMR 6299, Nantes, France
| | | | | | | | | | | | | |
Collapse
|
46
|
Wang J, Liao L, Wang S, Tan J. Cell therapy with autologous mesenchymal stem cells-how the disease process impacts clinical considerations. Cytotherapy 2013; 15:893-904. [PMID: 23751203 DOI: 10.1016/j.jcyt.2013.01.218] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 01/07/2013] [Accepted: 01/23/2013] [Indexed: 12/13/2022]
Abstract
The prospective clinical use of multipotent mesenchymal stromal cells (MSCs) holds enormous promise for the treatment of a large number of degenerative and age-related diseases. In particular, autologous MSCs isolated from bone marrow (BM) are considered safe and have been extensively evaluated in clinical trials. Nevertheless, different efficacies have been reported, depending on the health status and age of the donor. In addition, the biological functions of BM-MSCs from patients with various diseases may be impaired. Furthermore, medical treatments such as long-term chemotherapy and immunomodulatory therapy may damage the BM microenvironment and affect the therapeutic potential of MSCs. Therefore, a number of practical problems must be addressed before autologous BM-MSCs can be widely applied with higher efficiency in patients. As such, this review focuses on various factors that directly influence the biological properties of BM-MSCs, and we discuss the possible mechanisms of these alterations.
Collapse
Affiliation(s)
- Jin Wang
- Organ Transplant Institute, Fuzhou General Hospital, Xiamen University, Fuzhou, China
| | | | | | | |
Collapse
|
47
|
Gottschling S, Granzow M, Kuner R, Jauch A, Herpel E, Xu EC, Muley T, Schnabel PA, Herth FJF, Meister M. Mesenchymal stem cells in non-small cell lung cancer--different from others? Insights from comparative molecular and functional analyses. Lung Cancer 2013; 80:19-29. [PMID: 23294501 DOI: 10.1016/j.lungcan.2012.12.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/05/2012] [Accepted: 12/09/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAF) play a vital role in lung cancer initiation and progression. Although mesenchymal stem cells (MSC) are considered progenitor cells of fibroblasts and show cancer modulating abilities themselves, analyses on their presence and properties in lung cancer are lacking so far. METHODS We performed a comparative molecular and functional analysis of MSC derived from non-small cell lung cancer (NSCLC) and corresponding normal lung tissue (NLT) of a total of 15 patients. MSC were identified and selected according to their mesenchymal multilineage differentiation capability and surface marker profile. RESULTS Compared to NLT-MSC, NSCLC-MSC showed accelerated growth kinetics and reduced sensitivity to cisplatin. Karyotyping, comparative genomic hybridization and multiplex fluorescence in situ hybridization revealed no chromosomal aberrations. However, gene expression profiling of NSCLC- and NLT-MSC indicated variable expression of 62 genes involved in proliferation, DNA repair, apoptosis, extracellular matrix synthesis, tissue remodeling and angiogenesis. Differential expression of the selected candidate genes butyrylcholinesterase, clusterin and quiescin Q6 sulfhydryl oxidase 1 was validated by quantitative real-time PCR and, on protein level, by immunohistochemical analyses of original tumor tissue. Upon exposure to tumor cell-conditioned medium or transforming growth factor-β, both, NSCLC-MSC and NLT-MSC acquired expression of α-smooth muscle actin (α-SMA), a major characteristics of CAF. CONCLUSIONS This study indicates that NSCLC tissue contains MSC with specific molecular and functional properties. These cells might represent a progenitor reservoir for CAF and thus crucially contribute to lung cancer progression.
Collapse
Affiliation(s)
- Sandra Gottschling
- Department of Thoracic Oncology, Thoraxklinik, University of Heidelberg, Amalienstr. 5, 69126 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Etoposide and Hypoxia Do Not Activate Apoptosis of Multipotent Mesenchymal Stromal Cells In Vitro. Bull Exp Biol Med 2012; 154:141-4. [DOI: 10.1007/s10517-012-1895-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
49
|
Cruet-Hennequart S, Prendergast ÁM, Shaw G, Barry FP, Carty MP. Doxorubicin induces the DNA damage response in cultured human mesenchymal stem cells. Int J Hematol 2012; 96:649-56. [PMID: 23076878 DOI: 10.1007/s12185-012-1196-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 09/22/2012] [Accepted: 09/24/2012] [Indexed: 12/19/2022]
Abstract
Anthracyclines, including doxorubicin, are widely used in the treatment of leukemia. While the effects of doxorubicin on hematopoietic cells have been characterized, less is known about the response of human mesenchymal stem cells (hMSCs) in the bone marrow stroma to anthracyclines. We characterized the effect of doxorubicin on key DNA damage responses in hMSCs, and compared doxorubicin sensitivity and DNA damage response activation between isolated hMSCs and the chronic myelogenous leukemia cell line, K562. Phosphorylation of H2AX, Chk1, and RPA2 was more strongly activated in K562 cells than in hMSCs, at equivalent doses of doxorubicin. hMSCs were relatively resistant to doxorubicin such that, following exposure to 15 μM doxorubicin, the level of cleaved caspase-3 detected by western blotting was lower in hMSCs compared to K562 cells. Flow cytometric analysis of cell cycle progression demonstrated that exposure to doxorubicin induced G2/M phase arrest in hMSCs, while 48 h after exposure, 15.6 % of cells were apoptotic, as determined from the percentage of cells having sub-G1 DNA content. We also show that the doxorubicin sensitivity of hMSCs isolated from a healthy donor was comparable to that of hMSCs isolated from a chronic lymphocytic leukemia patient. Overall, our results demonstrate that high doses of doxorubicin induce the DNA damage response in hMSCs, and that cultured hMSCs are relatively resistant to doxorubicin.
Collapse
Affiliation(s)
- Séverine Cruet-Hennequart
- DNA Damage Response Laboratory, Centre for Chromosome Biology and School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| | | | | | | | | |
Collapse
|
50
|
Kucerova L, Poturnajova M, Tyciakova S, Matuskova M. Increased proliferation and chemosensitivity of human mesenchymal stromal cells expressing fusion yeast cytosine deaminase. Stem Cell Res 2012; 8:247-58. [DOI: 10.1016/j.scr.2011.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 10/14/2011] [Accepted: 11/25/2011] [Indexed: 01/14/2023] Open
|