1
|
Yurdakul-Mesutoglu P, Akin HY, Gunaydin Z. Ex Vivo Expansion of Cord Blood Hematopoietic Stem and Progenitor Cells. Methods Mol Biol 2025. [PMID: 40106144 DOI: 10.1007/7651_2025_610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Umbilical cord blood (CB)-derived hematopoietic stem and progenitor cells (HSPCs) hold immense potential for regenerative medicine, particularly in hematologic malignancies and immune disorders. CB offers several advantages, including easy collection and reduced risk of graft-versus-host disease compared to other sources, like bone marrow. However, the clinical application of CB is often limited due to the relatively small number of HSPCs present in CB grafts, which can be insufficient for adult patients. This limitation has prompted researchers to explore various methods to expand HSPCs ex vivo. As research continues to refine expansion techniques, the future of CB HSPC therapy appears increasingly promising, offering new hope for patients requiring stem cell transplantation. Approaches to HSPC expansion include the use of cytokines, small molecules, epigenetic modulators, and advanced culture systems that mimic the bone marrow niche as well as emerging techniques such as gene editing. Of the key CB HSPC expansion methodologies, the use of epigenetic modifiers is among the most promising strategies for inducing proliferation while maintaining the stemness of CB HSPC. This section summarizes key methodologies for CB HSPC expansion and their transformative impact on clinical practice while providing a validated protocol for ex vivo expansion of CB-derived HSPCs using valproic acid and/or nicotinamide.
Collapse
Affiliation(s)
| | - Hasan Yalim Akin
- Ankara University Faculty of Medicine, Cord Blood Bank, Ankara, Turkey
| | - Zeynep Gunaydin
- Ankara University Faculty of Medicine, Cord Blood Bank, Ankara, Turkey
| |
Collapse
|
2
|
Cui Y, Ren Y, Ren F, Zhang Y, Wang H. Synergistic effect and molecular mechanism of nicotinamide and UM171 in ex vivo expansion of long-term hematopoietic stem cells. Regen Ther 2024; 27:191-199. [PMID: 38840730 PMCID: PMC11150914 DOI: 10.1016/j.reth.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Several approaches to expand human hematopoietic stem cells (HSCs) have been reported, but the ability of these methods to expand long-term hematopoietic stem cells (LT-HSCs) remains to be improved, which limits the application of HSCs-based therapies. Methods CD34+ cells were purified from umbilical cord blood using MacsCD34 beads, and then cultured for 12 d in a serum-free medium. Flow cytometry was used to detect phenotype, cell cycle distribution, and apoptosis of the cultured cells. Colony-forming cell (CFC) assays can evaluate multi-lineage differentiation potential of HSCs. Real-time polymerase chain reaction was employed to detect the expression of genes related to self-renewal programs and antioxidant activity. DCFH-DA probes were used to evaluate intracellular production of reactive oxygen species (ROS). Determination of the effect of different culture conditions on the balance of cytokine by cytometric bead array. Results Here, we show a combination, Nicotinamide (NAM) combined with pyrimidoindole derivative UM171, can massively expand LT-HSCs ex vivo, and the expanded cells maintained the capability of self-renewal and multilineage differentiation. Additionally, our data indicated that UM171 promoted self-renewal of HSCs by inducing HSCs entry into the cell cycle and activating Notch and Wnt pathways, but the infinite occurrence of this process may lead to mitochondrial metabolism disorder and differentiation of HSCs. NAM kept HSCs in their primitive and dormant states by reducing intracellular ROS levels and upregulating the expression of stemness related genes, so we believed that NAM can act as a brake to control the above process. Conclusions The discovery of the synergistic effect of NAM and UM171 for expanding LT-HSCs provides a new strategy in solving the clinical issue of limited numbers of HSCs.
Collapse
Affiliation(s)
- Yanni Cui
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yan Ren
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Fanggang Ren
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Molecular Diagnosis and Treatment of Blood Diseases in Shanxi Province, Taiyuan, China
| | - Yaofang Zhang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Molecular Diagnosis and Treatment of Blood Diseases in Shanxi Province, Taiyuan, China
| | - Hongwei Wang
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Molecular Diagnosis and Treatment of Blood Diseases in Shanxi Province, Taiyuan, China
- Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
3
|
Gencer EB, Akin HY, Toprak SK, Turasan E, Yousefzadeh M, Yurdakul-Mesutoglu P, Cagan M, Seval MM, Katlan DC, Dalva K, Beksac MS, Beksac M. In vivo and in vitro effects of cord blood hematopoietic stem and progenitor cell (HSPC) expansion using valproic acid and/or nicotinamide. Curr Res Transl Med 2024; 72:103444. [PMID: 38447268 DOI: 10.1016/j.retram.2024.103444] [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: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND High self-renewal capacity and most permissive nature of umbilical cord blood (CB) results with successful transplant outcomes but low hematopoietic stem and progenitor cell (HSPC) counts limits wider use. In order to overcome this problem ex vivo expansion with small molecules such as Valproic acid (VPA) or Nicotinamide (NAM) have been shown to be effective. To the best of our knowledge, the combinatory effects of VPA and NAM on HSPC expansion has not been studied earlier. The aim of this study was to analyze ex vivo and in vivo efficacy of VPA and NAM either alone or in combination in terms of expansion and engraftment. METHODS A total of 44 CB units were included in this study. To determine the ex vivo and in vivo efficacy, human CB CD34+ cells were expanded with VPA and/or NAM and colony forming unit (CFU) assay was performed on expanded HSPC. Xenotransplantation was performed simultaneously by intravenous injection of expanded HSPC to NOD-SCID gamma (NSG) mice (n = 22). Significance of the difference between the expansion groups or xenotransplantation models was analyzed using t-test, Mann-Whitney, ANOVA or Kruskal-Wallis tests as appropriate considering the normality of distributions and the number of groups analyzed. RESULTS In vitro CD34+ HSPC expansion fold relative to cytokines-only was significantly higher with VPA compared to NAM [2.23 (1.07-5.59) vs 1.48 (1.00-4.40); p < 0.05]. Synergistic effect of VPA+NAM has achieved a maximum relative expansion fold at 21 days (D21) of incubation [2.95 (1.00-11.94)]. There was no significant difference between VPA and VPA+NAM D21 (p = 0.44). Fold number of colony-forming unit granulocyte-macrophage (CFU-GM) colonies relative to the cytokine-only group was in favor of NAM compared to VPA [1.87 (1.00-3.59) vs 1.00 (1.00-1.81); p < 0.01]. VPA+NAM D21 [1.62 (1.00-2.77)] was also superior against VPA (p < 0.05). There was no significant difference between NAM and VPA+NAM D21. Following human CB34+ CB transplantation (CBT) in the mouse model, fastest in vivo leukocyte recovery was observed with VPA+NAM expanded cells (6 ± 2 days) and the highest levels of human CD45 chimerism was detectable with VPA-expanded CBT (VPA: 5.42 % at day 28; NAM: 2.45 % at day 31; VPA+NAM 1.8 % at day 31). CONCLUSION Our study results suggest using VPA alone, rather than in combination with NAM or NAM alone, to achieve better and faster expansion and engraftment of CB HSPC.
Collapse
Affiliation(s)
| | - Hasan Yalim Akin
- Ankara University Faculty of Medicine Cord Blood Bank, Ankara, Turkey; Middle East Technical University, Department of Biochemistry, Ankara, Turkey
| | - Selami Kocak Toprak
- Ankara University Faculty of Medicine Cord Blood Bank, Ankara, Turkey; Ankara University Faculty of Medicine Department of Hematology, Ankara, Turkey
| | - Eylul Turasan
- Ankara University Faculty of Medicine Cord Blood Bank, Ankara, Turkey
| | - Mahsa Yousefzadeh
- Ankara University Faculty of Medicine Cord Blood Bank, Ankara, Turkey; Ankara University Stem Cell Institute, Ankara, Turkey
| | | | - Murat Cagan
- Hacettepe University Faculty of Medicine Department of Obstetrics and Gynecology, Ankara, Turkey
| | - Mehmet Murat Seval
- Ankara University Faculty of Medicine Department of Obstetrics and Gynecology, Ankara, Turkey
| | - Doruk Cevdi Katlan
- Istanbul Training and Research Hospital Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Klara Dalva
- Ankara University Stem Cell Institute, Ankara, Turkey
| | - Mehmet Sinan Beksac
- Hacettepe University Faculty of Medicine Department of Obstetrics and Gynecology, Ankara, Turkey; Istinye University, Ankara Liv Hospital, Obstetrics and Gynecology, Ankara, Turkey
| | - Meral Beksac
- Ankara University Faculty of Medicine Department of Hematology, Ankara, Turkey; Istinye University, Ankara Liv Hospital, Hematology and Stem Cell Transplantation Unit Ankara, Turkey.
| |
Collapse
|
4
|
Bastani S, Staal FJT, Canté-Barrett K. The quest for the holy grail: overcoming challenges in expanding human hematopoietic stem cells for clinical use. Stem Cell Investig 2023; 10:15. [PMID: 37457748 PMCID: PMC10345135 DOI: 10.21037/sci-2023-016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Hematopoietic stem cell (HSC) transplantation has been the golden standard for many hematological disorders. However, the number of HSCs obtained from several sources, including umbilical cord blood (UCB), often is insufficient for transplantation. For decades, maintaining or even expanding HSCs for therapeutic purposes has been a "holy grail" in stem cell biology. Different methods have been proposed to improve the efficiency of cell expansion and enhance homing potential such as co-culture with stromal cells or treatment with specific agents. Recent progress has shown that this is starting to become feasible using serum-free and well-defined media. Some of these protocols to expand HSCs along with genetic modification have been successfully applied in clinical trials and some others are studied in preclinical and clinical studies. However, the main challenges regarding ex vivo expansion of HSCs such as limited growth potential and tendency to differentiate in culture still need improvements. Understanding the biology of blood stem cells, their niche and signaling pathways has provided possibilities to regulate cell fate decisions and manipulate cells to optimize expansion of HSCs in vitro. Here, we review the plethora of HSC expansion protocols that have been proposed and indicate the current state of the art for their clinical application.
Collapse
Affiliation(s)
- Sepideh Bastani
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Kirsten Canté-Barrett
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Sun Z, Yao B, Xie H, Su X. Clinical Progress and Preclinical Insights Into Umbilical Cord Blood Transplantation Improvement. Stem Cells Transl Med 2022; 11:912-926. [PMID: 35972332 PMCID: PMC9492243 DOI: 10.1093/stcltm/szac056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/07/2022] [Indexed: 11/14/2022] Open
Abstract
The application of umbilical cord blood (UCB) as an important source of hematopoietic stem and progenitor cells (HSPCs) for hematopoietic reconstitution in the clinical context has steadily grown worldwide in the past 30 years. UCB has advantages that include rapid availability of donors, less strict HLA-matching demands, and low rates of graft-versus-host disease (GVHD) versus bone marrow (BM) and mobilized peripheral blood (PB). However, the limited number of HSPCs within a single UCB unit often leads to delayed hematopoietic engraftment, increased risk of transplant-related infection and mortality, and proneness to graft failure, thus hindering wide clinical application. Many strategies have been developed to improve UCB engraftment, most of which are based on 2 approaches: increasing the HSPC number ex vivo before transplantation and enhancing HSPC homing to the recipient BM niche after transplantation. Recently, several methods have shown promising progress in UCB engraftment improvement. Here, we review the current situations of UCB manipulation in preclinical and clinical settings and discuss challenges and future directions.
Collapse
Affiliation(s)
- Zhongjie Sun
- State Key Laboratory of Elemento-organic chemistry, College of Chemistry, Nankai University, Tianjin, People's Republic of China.,Newish Technology (Beijing) Co., Ltd., Beijing, People's Republic of China
| | - Bing Yao
- Zhejiang Hisoar Pharmaceutical Co., Ltd., Taizhou, Zhejiang Province, People's Republic of China
| | - Huangfan Xie
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, People's Republic of China.,Newish Technology (Beijing) Co., Ltd., Beijing, People's Republic of China
| | - XunCheng Su
- State Key Laboratory of Elemento-organic chemistry, College of Chemistry, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
6
|
Li Q, Hao S, Cheng T. [Research progress on in vitro expansion and clinical application of hematopoietic stem cell]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:167-172. [PMID: 35381684 PMCID: PMC8980649 DOI: 10.3760/cma.j.issn.0253-2727.2022.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Q Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - S Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - T Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
7
|
Li J, Wang X, Ding J, Zhu Y, Min W, Kuang W, Yuan K, Sun C, Yang P. Development and clinical advancement of small molecules for ex vivo expansion of hematopoietic stem cell. Acta Pharm Sin B 2021; 12:2808-2831. [PMID: 35755294 PMCID: PMC9214065 DOI: 10.1016/j.apsb.2021.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cell (HSC) transplantation is the only curative therapy for many diseases. HSCs from umbilical cord blood (UCB) source have many advantages over from bone marrow. However, limited HSC dose in a single CB unit restrict its widespread use. Over the past two decades, ex vivo HSC expansion with small molecules has been an effective approach for obtaining adequate HSCs. Till now, several small-molecule compounds have entered the phase I/II trials, showing safe and favorable pharmacological profiles. As HSC expansion has become a hot topic over recent years, many newly identified small molecules along with novel biological mechanisms for HSC expansion would help solve this challenging issue. Here, we will give an overview of HSC biology, discovery and medicinal chemistry development of small molecules, natural products targeting for HSC expansion, and their recent clinical progresses, as well as potential protein targets for HSC expansion.
Collapse
|
8
|
Feng Z, Lin C, Tu L, Su M, Song C, Liu S, Suryanto ME, Hsiao CD, Li L. FDA-Approved Drug Screening for Compounds That Facilitate Hematopoietic Stem and Progenitor Cells (HSPCs) Expansion in Zebrafish. Cells 2021; 10:cells10082149. [PMID: 34440919 PMCID: PMC8393331 DOI: 10.3390/cells10082149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a specialized subset of cells with self-renewal and multilineage differentiation potency, which are essential for their function in bone marrow or umbilical cord blood transplantation to treat blood disorders. Expanding the hematopoietic stem and progenitor cells (HSPCs) ex vivo is essential to understand the HSPCs-based therapies potency. Here, we established a screening system in zebrafish by adopting an FDA-approved drug library to identify candidates that could facilitate HSPC expansion. To date, we have screened 171 drugs of 7 categories, including antibacterial, antineoplastic, glucocorticoid, NSAIDS, vitamins, antidepressant, and antipsychotic drugs. We found 21 drugs that contributed to HSPCs expansion, 32 drugs’ administration caused HSPCs diminishment and 118 drugs’ treatment elicited no effect on HSPCs amplification. Among these drugs, we further investigated the vitamin drugs ergocalciferol and panthenol, taking advantage of their acceptability, limited side-effects, and easy delivery. These two drugs, in particular, efficiently expanded the HSPCs pool in a dose-dependent manner. Their application even mitigated the compromised hematopoiesis in an ikzf1−/− mutant. Taken together, our study implied that the larval zebrafish is a suitable model for drug repurposing of effective molecules (especially those already approved for clinical use) that can facilitate HSPCs expansion.
Collapse
Affiliation(s)
- Zhi Feng
- Key Laboratory of Freshwater Fish Reproduction and Development, Institute of Developmental Biology and Regenerative Medicine, Ministry of Education, Southwest University, Chongqing 400715, China; (Z.F.); (C.L.); (L.T.); (M.S.); (C.S.); (S.L.)
| | - Chenyu Lin
- Key Laboratory of Freshwater Fish Reproduction and Development, Institute of Developmental Biology and Regenerative Medicine, Ministry of Education, Southwest University, Chongqing 400715, China; (Z.F.); (C.L.); (L.T.); (M.S.); (C.S.); (S.L.)
| | - Limei Tu
- Key Laboratory of Freshwater Fish Reproduction and Development, Institute of Developmental Biology and Regenerative Medicine, Ministry of Education, Southwest University, Chongqing 400715, China; (Z.F.); (C.L.); (L.T.); (M.S.); (C.S.); (S.L.)
| | - Ming Su
- Key Laboratory of Freshwater Fish Reproduction and Development, Institute of Developmental Biology and Regenerative Medicine, Ministry of Education, Southwest University, Chongqing 400715, China; (Z.F.); (C.L.); (L.T.); (M.S.); (C.S.); (S.L.)
- Research Center of Stem Cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Chunyu Song
- Key Laboratory of Freshwater Fish Reproduction and Development, Institute of Developmental Biology and Regenerative Medicine, Ministry of Education, Southwest University, Chongqing 400715, China; (Z.F.); (C.L.); (L.T.); (M.S.); (C.S.); (S.L.)
| | - Shengnan Liu
- Key Laboratory of Freshwater Fish Reproduction and Development, Institute of Developmental Biology and Regenerative Medicine, Ministry of Education, Southwest University, Chongqing 400715, China; (Z.F.); (C.L.); (L.T.); (M.S.); (C.S.); (S.L.)
| | - Michael Edbert Suryanto
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
| | - Chung-Der Hsiao
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Center for Nanotechnology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Correspondence: (C.-D.H.); (L.L.)
| | - Li Li
- Research Center of Stem Cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Correspondence: (C.-D.H.); (L.L.)
| |
Collapse
|
9
|
Zimran E, Papa L, Hoffman R. Ex vivo expansion of hematopoietic stem cells: Finally transitioning from the lab to the clinic. Blood Rev 2021; 50:100853. [PMID: 34112560 DOI: 10.1016/j.blre.2021.100853] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 01/17/2023]
Abstract
Hematopoietic stem cells (HSCs) have been used for therapeutic purposes for decades in the form of autologous and allogeneic transplantation and are currently emerging as an attractive target for gene therapy. A low stem cell dose is a major barrier to the application of HSC therapy in several situations, primarily umbilical cord blood transplantation and gene modification. Strategies that promote ex vivo expansion of the numbers of functional HSCs could overcome this barrier, hence have been the subject of intense and prolonged research. Several ex vivo expansion strategies have advanced to evaluation clinical trials, which are showing favorable outcomes along with convincing safety signals. Preclinical studies have recently confirmed beneficial incorporation of ex vivo expansion into HSC gene modification protocols. Collectively, ex vivo HSC expansion holds promise for significantly broadening the availability of cord blood units for transplantation, and for optimizing gene therapy protocols to enable their clinical application.
Collapse
Affiliation(s)
- Eran Zimran
- Hematology Department, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Kiryat Hadassah 1, POB 1200, Jerusalem, 911200, Israel.
| | - Luena Papa
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, Box 1079, New York, NY 10029, USA.
| | - Ronald Hoffman
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, Box 1079, New York, NY 10029, USA.
| |
Collapse
|
10
|
Sica RA, Terzioglu MK, Mahmud D, Mahmud N. Mechanistic Basis of ex Vivo Umbilical Cord Blood Stem Progenitor Cell Expansion. Stem Cell Rev Rep 2021; 16:628-638. [PMID: 32424674 DOI: 10.1007/s12015-020-09981-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Umbilical cord blood (CB) transplantation has been used successfully in humans for three decades due to its rapid availability for patients lacking a suitable allogeneic donor, less stringent HLA matching requirements, and low rates of relapse and chronic graft-versus-host disease (GVHD). However, CB transplantation is associated with complications, such as delayed hematopoietic engraftment, graft failure, which increases infection and bleeding and causes longer hospital stays, and transplant-related mortality. The majority of these biological limitations are due to the unforeseeable functional potency of multipotent hematopoietic stem cells (HSCs), which reduce the predictability of successful transplantation; however, several strategies have been developed to increase the number of hematopoietic stem progenitor cells (HSPCs) infused during CB transplantation. This review primarily addresses the methods that promote ex vivo CB expansion within the context of symmetrical and asymmetrical HSC division and those that rely on epigenetic mechanisms, along with the reportedly most successful cytokine combinations. We also review recent clinical research on small molecules (StemRegenin-1, UM171, and nicotinamide) in ex vivo expanded CB and discuss yet unvalidated preclinical strategies. Expanding and transplanting CB graft enriched in HSPCs in a single CB unit is a particularly exciting prospect with the potential to improve the use and availability of CB grafts. Greater knowledge of optimal ex vivo expansion strategies, cell longevity, and graft potency will expand the scope of cellular therapies. Also the development of adequate ex vivo HSPC expansion strategies could bring expanded cord blood grafts to the forefront of transplant therapy and regenerative medicine.
Collapse
Affiliation(s)
- R Alejandro Sica
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA
| | - Meryem K Terzioglu
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA
| | - Dolores Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA
| | - Nadim Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
11
|
Ghafouri-Fard S, Niazi V, Taheri M, Basiri A. Effect of Small Molecule on ex vivo Expansion of Cord Blood Hematopoietic Stem Cells: A Concise Review. Front Cell Dev Biol 2021; 9:649115. [PMID: 33898442 PMCID: PMC8063724 DOI: 10.3389/fcell.2021.649115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a group of cells being produced during embryogenesis to preserve the blood system. They might also be differentiated to non-hematopoietic cells, including neural, cardiac and myogenic cells. Therefore, they have vast applications in the treatment of human disorders. Considering the restricted quantities of HSCs in the umbilical cord blood, inadequate mobilization of bone marrow stem cells, and absence of ethnic dissimilarity, ex vivo expansion of these HSCs is an applicable method for obtaining adequate amounts of HSCs. Several molecules such as NR-101, zVADfmk, zLLYfmk, Nicotinamide, Resveratrol, the Copper chelator TEPA, dmPGE2, Garcinol, and serotonin have been used in combination of cytokines to expand HSCs ex vivo. The most promising results have been obtained from cocktails that influence multipotency and self-renewal features from different pathways. In the current manuscript, we provide a concise summary of the effects of diverse small molecules on expansion of cord blood HSCs.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Zarrabi M, Afzal E, Asghari MH, Ebrahimi M. Combination of SB431542, Chir9901, and Bpv as a novel supplement in the culture of umbilical cord blood hematopoietic stem cells. Stem Cell Res Ther 2020; 11:474. [PMID: 33168035 PMCID: PMC7650159 DOI: 10.1186/s13287-020-01945-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/20/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Small molecule compounds have been well recognized for their promising power in the generation, expansion, and maintenance of embryonic or adult stem cells. The aim of this study was to identify a novel combination of small molecules in order to optimize the ex vivo expansion of umbilical cord blood-derived CD34+ cells. METHODS Considering the most important signaling pathways involved in the self-renewal of hematopoietic stem cells, CB-CD34+ cells were expanded with cytokines in the presence of seven small molecules including SB, PD, Chir, Bpv, Pur, Pμ, and NAM. The eliminativism approach was used to find the best combination of selected small molecules for effective ex vivo expansion of CD34+ cell. In each step, proliferation, self-renewal, and clonogenic potential of the expanded cells as well as expression of some hematopoietic stem cell-related genes were studied. Finally, the engraftment potential of expanded cells was also examined by the mouse intra-uterine transplantation model. RESULTS Our data shows that the simultaneous use of SB431542 (TGF-β inhibitor), Chir9901 (GSK3 inhibitor), and Bpv (PTEN inhibitor) resulted in a 50-fold increase in the number of CD34+CD38- cells. This was further reflected in approximately 3 times the increase in the clonogenic potential of the small molecule cocktail-expanded cells. These cells, also, showed a 1.5-fold higher engraftment potential in the peripheral blood of the NMRI model of in utero transplantation. These results are in total conformity with the upregulation of HOXB4, GATA2, and CD34 marker gene as well as the CXCR4 homing gene. CONCLUSION Taken together, our findings introduce a novel combination of small molecules to improve the yield of existing protocols used in the expansion of hematopoietic stem cells.
Collapse
Affiliation(s)
- Morteza Zarrabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box, Tehran, 19395-4644, Iran
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Elaheh Afzal
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Mohammad Hassan Asghari
- Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Animal Biotechnology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box, Tehran, 19395-4644, Iran.
| |
Collapse
|
13
|
Zaccara IM, Mestieri LB, Pilar EFS, Moreira MS, Grecca FS, Martins MD, Kopper PMP. Photobiomodulation therapy improves human dental pulp stem cell viability and migration in vitro associated to upregulation of histone acetylation. Lasers Med Sci 2020; 35:741-749. [PMID: 32095920 DOI: 10.1007/s10103-019-02931-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 11/27/2019] [Indexed: 12/17/2022]
Abstract
This in vitro study evaluated the role of photobiomodulation therapy (PBMT) on viability and migration of human dental pulp stem cells (hDPSCs) and its association to epigenetic mechanisms such as histone acetylation. The hDPSCs were characterized and assigned into control and PBMT groups. For the PBMT, five laser irradiations at 6-h intervals were performed using a continuous-wave InGaAlP diode laser. Viability (MTT), migration (scratch), and histone acetylation H3 (H3K9ac immunofluorescence) were evaluated immediately after the last irradiation. PBMT significantly increased the viability (P = 0.004). Also, PBMT group showed significantly increased migration of cells in the wound compared to the control in 6 h (P = 0.002), 12 h (P = 0.014) and 18 h (P = 0.083) being faster than the control, which only finished the process at 24 h. PBMT induced epigenetic modifications in hDPSC due to increased histone acetylation (P = 0.001). PBMT increased viability and migration of hDPSCs, which are related with the upregulation of histone acetylation and could be considered a promising adjuvant therapy for regenerative endodontic treatment.
Collapse
Affiliation(s)
- Ivana M Zaccara
- Dentistry Graduate Program, Department of Conservative Dentistry, School of Dentistry, Federal University of Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Letícia B Mestieri
- Dentistry Graduate Program, Department of Conservative Dentistry, School of Dentistry, Federal University of Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Emily F S Pilar
- Department of Experimental Pathology, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Maria S Moreira
- Department of Dentistry, School of Dentistry, University of Sao Paulo, Sao Paulo, Brazil.,Ibirapuera University, Sao Paulo, Brazil
| | - Fabiana S Grecca
- Dentistry Graduate Program, Department of Conservative Dentistry, School of Dentistry, Federal University of Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Manoela D Martins
- Dentistry Graduate Program, Department of Conservative Dentistry, School of Dentistry, Federal University of Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Patrícia Maria Poli Kopper
- Dentistry Graduate Program, Department of Conservative Dentistry, School of Dentistry, Federal University of Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2492, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.
| |
Collapse
|
14
|
Zimran E, Papa L, Djedaini M, Patel A, Iancu-Rubin C, Hoffman R. Expansion and preservation of the functional activity of adult hematopoietic stem cells cultured ex vivo with a histone deacetylase inhibitor. Stem Cells Transl Med 2020; 9:531-542. [PMID: 31950644 PMCID: PMC7103619 DOI: 10.1002/sctm.19-0199] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/27/2019] [Indexed: 12/17/2022] Open
Abstract
Attempts to expand ex vivo the numbers of human hematopoietic stem cells (HSCs) without compromising their marrow repopulating capacity and their ability to establish multilineage hematopoiesis has been the subject of intense investigation. Although most such efforts have focused on cord blood HSCs, few have been applied to adult HSCs, a more clinically relevant HSC source for gene modification. To date, the strategies that have been used to expand adult HSCs have resulted in modest effects or HSCs with lineage bias and a limited ability to generate T cells in vivo. We previously reported that culturing umbilical cord blood CD34+ cells in serum‐free media supplemented with valproic acid (VPA), a histone deacetylase inhibitor, and a combination of cytokines led to the expansion of the numbers of fully functional HSCs. In the present study, we used this same approach to expand the numbers of adult human CD34+ cells isolated from mobilized peripheral blood and bone marrow. This approach resulted in a significant increase in the numbers of phenotypically defined HSCs (CD34+CD45RA‐CD90+D49f+). Cells incubated with VPA also exhibited increased aldehyde dehydrogenase activity and decreased mitochondrial membrane potential, each functional markers of HSCs. Grafts harvested from VPA‐treated cultures were able to engraft in immune‐deficient mice and, importantly, to generate cellular progeny belonging to each hematopoietic lineage in similar proportion to that observed with unmanipulated CD34+ cells. These data support the utility of VPA‐mediated ex vivo HSC expansion for gene modification of adult HSCs.
Collapse
Affiliation(s)
- Eran Zimran
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.,Hematology Department, Hadassah University Center, Jerusalem, Israel
| | - Luena Papa
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mansour Djedaini
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ami Patel
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Camelia Iancu-Rubin
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ronald Hoffman
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
15
|
Bari S, Chong P, Hwang WYK. Expansion of Haematopoietic Stem and Progenitor Cells: Paving the Way for Next-Generation Haematopoietic Stem Cell Transplantation. BLOOD CELL THERAPY 2019; 2:58-67. [PMID: 37588101 PMCID: PMC10427230 DOI: 10.31547/bct-2019-004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/18/2019] [Indexed: 08/18/2023]
Abstract
Haematopoietic stem cell transplantation (HSCT) is now an established practice with over 70,000 transplants performed annually, and over 1.5 million around the world so far. The practice of HSCT has improved over the years due to advances in conditioning regiments, preparatory practices for patients leading up to the transplant, graft versus host disease (GVHD) and infection prophylaxis, as well as a better selection of patients. However, in many instances, the stem cells supplied to the patient may not be adequate for optimal transplantation outcomes. This may be seen in a few areas including umbilical cord blood transplantation, inadequate bone marrow, peripheral blood stem cell harvest, or gene therapy. Growing and expanding HSCs in culture would provide an increase in cell numbers prior to stem cell infusion and accelerate haematopoietic recovery, resulting in improved outcomes. Several new technologies have emerged in recent years, which have facilitated the expansion of haematopoietic stem and progenitor cells (HSPCs) in culture with good outcomes in vitro, in vivo, and in clinical trials. In this review, we will outline some of the reasons for the expansion of HSPCs as well as the new technologies facilitating the advances in HSCT.
Collapse
Affiliation(s)
- Sudipto Bari
- National Cancer Centre Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | | | - William Ying Khee Hwang
- National Cancer Centre Singapore
- Department of Haematology, Singapore General Hospital, Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| |
Collapse
|
16
|
Ex vivo human HSC expansion requires coordination of cellular reprogramming with mitochondrial remodeling and p53 activation. Blood Adv 2019; 2:2766-2779. [PMID: 30348672 DOI: 10.1182/bloodadvances.2018024273] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/06/2018] [Indexed: 01/02/2023] Open
Abstract
The limited number of hematopoietic stem cells (HSCs) in umbilical cord blood (UCB) units restricts their use for stem cell transplantation. Ex vivo treatment of UCB-CD34+ cells with valproic acid (VPA) increases the number of transplantable HSCs. In this study, we demonstrate that HSC expansion is not merely a result of proliferation of the existing stem cells but, rather, a result of a rapid reprogramming of CD34+CD90- cells into CD34+CD90+ cells, which is accompanied by limited numbers of cell divisions. Beyond this phenotypic switch, the treated cells acquire and retain a transcriptomic and mitochondrial profile, reminiscent of primary HSCs. Single and bulk RNA-seq revealed a signature highly enriched for transcripts characteristic of primary HSCs. The acquisition of this HSC signature is linked to mitochondrial remodeling accompanied by a reduced activity and enhanced glycolytic potential. These events act in concert with a modest upregulation of p53 activity to limit the levels of reactive oxygen species (ROS). Inhibition of either glycolysis or p53 activity impairs HSC expansion. This study indicates that a complex interplay of events is required for effective ex vivo expansion of UCB-HSCs.
Collapse
|
17
|
Tatetsu H, Armant M, Wang F, Gao C, Ueno S, Tian X, Federation A, Qi J, Bradner J, Tenen DG, Chai L. Maintenance and enhancement of human peripheral blood mobilized stem/progenitor cell engraftment after ex vivo culture via an HDACi/SALL4 axis (3465). Exp Hematol 2019; 75:53-63.e11. [PMID: 31260717 DOI: 10.1016/j.exphem.2019.06.473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023]
Abstract
Currently, there is a growing need for culturing hematopoietic stem/progenitor cells (HSPCs) in vitro for various clinical applications including gene therapy. Compared with cord blood (CB) CD34+ HSPCs, it is more challenging to maintain or expand CD34+ peripheral blood mobilized stem/progenitor cells (PBSCs) ex vivo. To fill this knowledge gap, we have systematically surveyed 466 small-molecule drug compounds for their potential in cytokine-dependent expansion of human CD34+CD90+ HSPCs. We found that epigenetic modifiers, especially histone deacetylase inhibitors (HDACis), could preferentially maintain and expand these cells. In particular, treatment of CD34+ PBSCs with a single dose of HDACi trichostatin A (TSA) at a concentration of 50 nmol/L ex vivo yielded the greatest expansion (11.7-fold) of CD34+CD90+ cells when compared with the control (dimethyl sulfoxide [DMSO] plus cytokines) group. Additionally, TSA-treated PBSC CD34+ cells had a statistically significant higher engraftment rate than the control-treated group in xenotransplantation experiments. Mechanistically, TSA treatment was associated with increased expression of HSPC-related genes such as GATA2 and SALL4. Furthermore, TSA-mediated CD34+CD90+ expansion was reduced by downregulation of SALL4 but not GATA2. Overall, we have developed a robust, short-term (5-day), PBSC ex vivo maintenance/expansion culture technique and found that the HDACi-TSA/SALL4 axis is important for the biological process.
Collapse
Affiliation(s)
- Hiro Tatetsu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Myriam Armant
- Trans Lab, Children's Hospital Boston, Boston, Massachusetts
| | - Fei Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shikiko Ueno
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xi Tian
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alex Federation
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jun Qi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore Centre for Translational Medicine, Singapore; Harvard Stem Cell Institute, Boston, Massachusetts
| | - Li Chai
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
18
|
Derakhshani M, Abbaszadeh H, Movassaghpour AA, Mehdizadeh A, Ebrahimi-Warkiani M, Yousefi M. Strategies for elevating hematopoietic stem cells expansion and engraftment capacity. Life Sci 2019; 232:116598. [PMID: 31247209 DOI: 10.1016/j.lfs.2019.116598] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/22/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cells (HSCs) are a rare cell population in adult bone marrow, mobilized peripheral blood, and umbilical cord blood possessing self-renewal and differentiation capability into a full spectrum of blood cells. Bone marrow HSC transplantation has been considered as an ideal option for certain disorders treatment including hematologic diseases, leukemia, immunodeficiency, bone marrow failure syndrome, genetic defects such as thalassemia, sickle cell anemia, autoimmune disease, and certain solid cancers. Ex vivo proliferation of these cells prior to transplantation has been proposed as a potential solution against limited number of stem cells. In such culture process, MSCs have also been shown to exhibit high capacity for secretion of soluble mediators contributing to the principle biological and therapeutic activities of HSCs. In addition, endothelial cells have been introduced to bridge the blood and sub tissues in the bone marrow, as well as, HSCs regeneration induction and survival. Cell culture in the laboratory environment requires cell growth strict control to protect against contamination, symmetrical cell division and optimal conditions for maximum yield. In this regard, microfluidic systems provide culture and analysis capabilities in micro volume scales. Moreover, two-dimensional cultures cannot fully demonstrate extracellular matrix found in different tissues and organs as an abstract representation of three dimensional cell structure. Microfluidic systems can also strongly describe the effects of physical factors such as temperature and pressure on cell behavior.
Collapse
Affiliation(s)
- Mehdi Derakhshani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Abbaszadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ebrahimi-Warkiani
- School of Biomedical Engineering, University Technology of Sydney, Sydney, New South Wales, 2007, Australia
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Hua P, Kronsteiner B, van der Garde M, Ashley N, Hernandez D, Tarunina M, Hook L, Choo Y, Roberts I, Mead A, Watt SM. Single-cell assessment of transcriptome alterations induced by Scriptaid in early differentiated human haematopoietic progenitors during ex vivo expansion. Sci Rep 2019; 9:5300. [PMID: 30923342 PMCID: PMC6438964 DOI: 10.1038/s41598-019-41803-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/18/2019] [Indexed: 12/24/2022] Open
Abstract
Priming haematopoietic stem/progenitor cells (HSPCs) in vitro with specific chromatin modifying agents and cytokines under serum-free-conditions significantly enhances engraftable HSC numbers. We extend these studies by culturing human CD133+ HSPCs on nanofibre scaffolds to mimic the niche for 5-days with the HDAC inhibitor Scriptaid and cytokines. Scriptaid increases absolute Lin−CD34+CD38−CD45RA−CD90+CD49f+ HSPC numbers, while concomitantly decreasing the Lin−CD38−CD34+CD45RA−CD90− subset. Hypothesising that Scriptaid plus cytokines expands the CD90+ subset without differentiation and upregulates CD90 on CD90− cells, we sorted, then cultured Lin−CD34+CD38−CD45RA−CD90− cells with Scriptaid and cytokines. Within 2-days and for at least 5-days, most CD90− cells became CD90+. There was no significant difference in the transcriptomic profile, by RNAsequencing, between cytokine-expanded and purified Lin−CD34+CD38−CD45RA−CD49f+CD90+ cells in the presence or absence of Scriptaid, suggesting that Scriptaid maintains stem cell gene expression programs despite expansion in HSC numbers. Supporting this, 50 genes were significantly differentially expressed between CD90+ and CD90− Lin−CD34+CD38−CD45RA−CD49f+ subsets in Scriptaid-cytokine- and cytokine only-expansion conditions. Thus, Scriptaid treatment of CD133+ cells may be a useful approach to expanding the absolute number of CD90+ HSC, without losing their stem cell characteristics, both through direct effects on HSC and potentially also conversion of their immediate CD90− progeny into CD90+ HSC.
Collapse
Affiliation(s)
- Peng Hua
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.,Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ, UK
| | - Barbara Kronsteiner
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ, UK
| | - Mark van der Garde
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ, UK
| | - Neil Ashley
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Diana Hernandez
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, SG1 2FX, UK
| | - Marina Tarunina
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, SG1 2FX, UK
| | - Lilian Hook
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, SG1 2FX, UK
| | - Yen Choo
- Plasticell Ltd, Stevenage Bioscience Catalyst, Stevenage, SG1 2FX, UK
| | - Irene Roberts
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.,Department of Paediatrics, University of Oxford, Children's Hospital, John Radcliffe Hospital, Oxford, OX3 9DU, UK.,Haematology Theme, Oxford Biomedical Research Centre, Oxford University Hospitals, Oxford, UK
| | - Adam Mead
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.,Haematology Theme, Oxford Biomedical Research Centre, Oxford University Hospitals, Oxford, UK
| | - Suzanne M Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and NHS Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ, UK.
| |
Collapse
|
20
|
Petro B, Mahmud D, Taioli S, Ganapathy A, Senyuk V, Yoshinaga KG, Suphangul M, Rondelli D, Mahmud N. Chromatin-Modifying Agent-Expanded Human Cord Blood Cells Display Reduced Allostimulatory Capacity. THE JOURNAL OF IMMUNOLOGY 2019; 202:2493-2501. [PMID: 30842275 DOI: 10.4049/jimmunol.1800128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 01/31/2019] [Indexed: 12/20/2022]
Abstract
The limited number of hematopoietic stem cells (HSC) within a single unit of human cord blood currently limits its use as an alternate graft source. However, we have developed a strategy using 5-aza-2'-deoxycytidine (5azaD) and trichostatin A (TSA), which expands transplantable HSC 7- to 10-fold. In our current studies, we have assessed the allostimulatory capacity of the 5azaD/TSA-expanded grafts. The coexpression of immunophenotypic dendritic cell (DC) markers, such as HLA-DR/CD86 and HLA-DR/CD11c as determined by flow cytometry, and the allostimulatory capacity of 5azaD/TSA-expanded cells as determined by MLC were both significantly lower than control. It has been previously demonstrated that STAT3 is indispensable for the differentiation of DC from HSC. Real-time quantitative PCR analysis revealed that 5azaD/TSA-expanded cells expressed more STAT3 transcript than control while also expressing increased transcripts for STAT3 inhibitors including SHP1, p21, and GATA1. Western blot analysis indicates that chromatin-modifying agent-expanded grafts displayed a reduced ratio of p-STAT3 to total STAT3 than control cultures, which is likely indicative of STAT3 inactivity in 5azD/TSA-expanded grafts. Culturing 5azaD/TSA-expanded cord blood cells in extended cultures reveals that they are still capable of generating DC. Notably, STAT3 inactivity was transient because the transcript levels of STAT3 and its inhibitors, including SHP1, were comparable between 5azaD/TSA and control cultures following extended culture. Taken together, our studies indicate that the reduced allostimulatory capacity of 5azaD/TSA-expanded cells is likely because of reversible inhibition of STAT3-dependent DC differentiation. These results suggest that a graft composed of 5azaD/TSA-expanded cells possesses relatively less allostimulatory response but is still capable of generating DC in permissive conditions.
Collapse
Affiliation(s)
- Benjamin Petro
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Dolores Mahmud
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Simona Taioli
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Amudha Ganapathy
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Vitalyi Senyuk
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Kazumi G Yoshinaga
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Montha Suphangul
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and
| | - Damiano Rondelli
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and.,University of Illinois Cancer Center, Chicago, IL 60612
| | - Nadim Mahmud
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612; and .,University of Illinois Cancer Center, Chicago, IL 60612
| |
Collapse
|
21
|
Zarrabi M, Afzal E, Ebrahimi M. Manipulation of Hematopoietic Stem Cell Fate by Small Molecule Compounds. Stem Cells Dev 2018; 27:1175-1190. [DOI: 10.1089/scd.2018.0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Morteza Zarrabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Elaheh Afzal
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
22
|
Zhang C, Zhang E, Yang L, Tu W, Lin J, Yuan C, Bunpetch V, Chen X, Ouyang H. Histone deacetylase inhibitor treated cell sheet from mouse tendon stem/progenitor cells promotes tendon repair. Biomaterials 2018; 172:66-82. [PMID: 29723756 DOI: 10.1016/j.biomaterials.2018.03.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/17/2018] [Accepted: 03/25/2018] [Indexed: 12/13/2022]
Abstract
Tendon stem/progenitor cells (TSPCs) have been identified as a rare population in tendons. In vitro propagation is indispensable to obtain sufficient quantities of TSPCs for therapies. However, culture-expanded TSPCs are prone to lose their phenotype, resulting in an inferior repaired capability. And little is known about the underlying mechanism. Here, we found that altered gene expression was associated with increased histone deacetylase (HDAC) activity and expression of HDAC subtypes. Therefore, we exposed ScxGFP mice-derived TSPCs to HDAC inhibitor (HDACi) trichostatin A (TSA) or valproic acid (VPA), and observed significant expansion of ScxGFP+ cells without altering phenotypic properties. TSA upregulated Scx expression by inhibiting HDAC1 and -3, and increasing the H3K27Ac level of Tgfb1 and -2 genome region. Additionally, cell sheets formed from TSA-pretreated mTSPCs retained the ability to accelerate tendon repair in vivo. Thus, our results uncovered an unrecognized role of HDACi in phenotypic and functional mTSPCs expansion to enhance their therapeutic potential.
Collapse
Affiliation(s)
- Can Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China; Institute of Biomedical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Erchen Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Long Yang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Wenjing Tu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Chunhui Yuan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Varisara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China.
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China; Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou 310058, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
23
|
Bari S, Zhong Q, Fan X, Poon Z, Lim AST, Lim TH, Dighe N, Li S, Chiu GNC, Chai CLL, Hwang WYK. Ex Vivo Expansion of CD34 + CD90 + CD49f + Hematopoietic Stem and Progenitor Cells from Non-Enriched Umbilical Cord Blood with Azole Compounds. Stem Cells Transl Med 2018; 7:376-393. [PMID: 29392885 PMCID: PMC5905230 DOI: 10.1002/sctm.17-0251] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/28/2017] [Indexed: 12/14/2022] Open
Abstract
Umbilical cord blood (UCB) transplants in adults have slower hematopoietic recovery compared to bone marrow (BM) or peripheral blood (PB) stem cells mainly due to low number of total nucleated cells and hematopoietic stem and progenitor cells (HSPC). As such in this study, we aimed to perform ex vivo expansion of UCB HSPC from non-enriched mononucleated cells (MNC) using novel azole-based small molecules. Freshly-thawed UCB-MNC were cultured in expansion medium supplemented with small molecules and basal cytokine cocktail. The effects of the expansion protocol were measured based on in vitro and in vivo assays. The proprietary library of >50 small molecules were developed using structure-activity-relationship studies of SB203580, a known p38-MAPK inhibitor. A particular analog, C7, resulted in 1,554.1 ± 27.8-fold increase of absolute viable CD45+ CD34+ CD38- CD45RA- progenitors which was at least 3.7-fold higher than control cultures (p < .001). In depth phenotypic analysis revealed >600-fold expansion of CD34+ /CD90+ /CD49f+ rare HSPCs coupled with significant (p < .01) increase of functional colonies from C7 treated cells. Transplantation of C7 expanded UCB grafts to immunodeficient mice resulted in significantly (p < .001) higher engraftment of human CD45+ and CD45+ CD34+ cells in the PB and BM by day 21 compared to non-expanded and cytokine expanded grafts. The C7 expanded grafts maintained long-term human multilineage chimerism in the BM of primary recipients with sustained human CD45 cell engraftment in secondary recipients. In conclusion, a small molecule, C7, could allow for clinical development of expanded UCB grafts without pre-culture stem cell enrichment that maintains in vitro and in vivo functionality. Stem Cells Translational Medicine 2018;7:376-393.
Collapse
Affiliation(s)
- Sudipto Bari
- Department of Hematology, Singapore General Hospital, Singapore, Singapore.,Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Qixing Zhong
- Department of Pharmacy, National University of Singapore, Singapore, Singapore.,Genome Institute of Singapore, Agency for Science and Technology Research, Singapore, Singapore
| | - Xiubo Fan
- Department of Clinical Research, Singapore General Hospital, Singapore, Singapore
| | - Zhiyong Poon
- Department of Hematology, Singapore General Hospital, Singapore, Singapore
| | | | - Tse Hui Lim
- Molecular Pathology, Singapore General Hospital, Singapore, Singapore
| | - Niraja Dighe
- Department of Hematology, Singapore General Hospital, Singapore, Singapore
| | - Shang Li
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Gigi Ngar Chee Chiu
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | | | - William Ying Khee Hwang
- Department of Hematology, Singapore General Hospital, Singapore, Singapore.,Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.,National Cancer Center Singapore, Singapore, Singapore.,Singapore Cord Blood Bank, Singapore, Singapore
| |
Collapse
|
24
|
Arulmozhivarman G, Kräter M, Wobus M, Friedrichs J, Bejestani EP, Müller K, Lambert K, Alexopoulou D, Dahl A, Stöter M, Bickle M, Shayegi N, Hampe J, Stölzel F, Brand M, von Bonin M, Bornhäuser M. Zebrafish In-Vivo Screening for Compounds Amplifying Hematopoietic Stem and Progenitor Cells: - Preclinical Validation in Human CD34+ Stem and Progenitor Cells. Sci Rep 2017; 7:12084. [PMID: 28935977 PMCID: PMC5608703 DOI: 10.1038/s41598-017-12360-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/08/2017] [Indexed: 01/13/2023] Open
Abstract
The identification of small molecules that either increase the number and/or enhance the activity of human hematopoietic stem and progenitor cells (hHSPCs) during ex vivo expansion remains challenging. We used an unbiased in vivo chemical screen in a transgenic (c-myb:EGFP) zebrafish embryo model and identified histone deacetylase inhibitors (HDACIs), particularly valproic acid (VPA), as significant enhancers of the number of phenotypic HSPCs, both in vivo and during ex vivo expansion. The long-term functionality of these expanded hHSPCs was verified in a xenotransplantation model with NSG mice. Interestingly, VPA increased CD34+ cell adhesion to primary mesenchymal stromal cells and reduced their in vitro chemokine-mediated migration capacity. In line with this, VPA-treated human CD34+ cells showed reduced homing and early engraftment in a xenograft transplant model, but retained their long-term engraftment potential in vivo, and maintained their differentiation ability both in vitro and in vivo. In summary, our data demonstrate that certain HDACIs lead to a net expansion of hHSPCs with retained long-term engraftment potential and could be further explored as candidate compounds to amplify ex-vivo engineered peripheral blood stem cells.
Collapse
Affiliation(s)
| | - Martin Kräter
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Manja Wobus
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Jens Friedrichs
- Institute of Biofunctional Polymer Materials, Leibniz Institute for Polymer Research, Max Bergmann Center of Biomaterials, Dresden, Germany
| | - Elham Pishali Bejestani
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site, Dresden, Germany
| | - Katrin Müller
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Katrin Lambert
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Dimitra Alexopoulou
- Deep Sequencing Group SFB655, Biotechnology Center, Technical University of Dresden, Dresden, Germany
| | - Andreas Dahl
- Deep Sequencing Group SFB655, Biotechnology Center, Technical University of Dresden, Dresden, Germany
| | - Martin Stöter
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marc Bickle
- Max-Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nona Shayegi
- Department of Hematology, University Hospital Essen, University of Duisburg, Essen, Germany
| | - Jochen Hampe
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Friedrich Stölzel
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany
| | - Michael Brand
- DFG-Center for Regenerative Therapies Dresden (CRTD) - Cluster of Excellence, Technical University of Dresden, Dresden, Germany.
| | - Malte von Bonin
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), partner site, Dresden, Germany
| | - Martin Bornhäuser
- Department of Hematology/Oncology, Medical Clinic and Policlinic I, University Hospital, Dresden, Germany. .,DFG-Center for Regenerative Therapies Dresden (CRTD) - Cluster of Excellence, Technical University of Dresden, Dresden, Germany.
| |
Collapse
|
25
|
Luo Z, Wang Z, He X, Liu N, Liu B, Sun L, Wang J, Ma F, Duncan H, He W, Cooper P. Effects of histone deacetylase inhibitors on regenerative cell responses in human dental pulp cells. Int Endod J 2017; 51:767-778. [DOI: 10.1111/iej.12779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 03/30/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Z. Luo
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
- Department of Operative Dentistry and Endodontics; School of Stomatology; The Guizhou Medical University; Guiyang China
| | - Z. Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - X. He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - N. Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - B. Liu
- Department of Stomatology; the Lishilu out-patient Department of the Chinese PLA Second Artillery Corps; Beijing China
| | - L. Sun
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - J. Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - F. Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - H. Duncan
- Division of Restorative Dentistry and Periodontology; Dublin Dental University Hospital; Dublin Ireland
| | - W. He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shanxi Key Laboratory of Stomatology; Department of Operative Dentistry and Endodontics; School of Stomatology; The Fourth Military Medical University; Xi'an China
| | - P. Cooper
- Oral Biology; School of Dentistry; University of Birmingham; Birmingham UK
| |
Collapse
|
26
|
Duncan HF, Smith AJ, Fleming GJP, Partridge NC, Shimizu E, Moran GP, Cooper PR. The Histone-Deacetylase-Inhibitor Suberoylanilide Hydroxamic Acid Promotes Dental Pulp Repair Mechanisms Through Modulation of Matrix Metalloproteinase-13 Activity. J Cell Physiol 2017; 231:798-816. [PMID: 26264761 DOI: 10.1002/jcp.25128] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/23/2022]
Abstract
Direct application of histone-deacetylase-inhibitors (HDACis) to dental pulp cells (DPCs) induces chromatin changes, promoting gene expression and cellular-reparative events. We have previously demonstrated that HDACis (valproic acid, trichostatin A) increase mineralization in dental papillae-derived cell-lines and primary DPCs by stimulation of dentinogenic gene expression. Here, we investigated novel genes regulated by the HDACi, suberoylanilide hydroxamic acid (SAHA), to identify new pathways contributing to DPC differentiation. SAHA significantly compromised DPC viability only at relatively high concentrations (5 μM); while low concentrations (1 μM) SAHA did not increase apoptosis. HDACi-exposure for 24 h induced mineralization-per-cell dose-dependently after 2 weeks; however, constant 14d SAHA-exposure inhibited mineralization. Microarray analysis (24 h and 14 days) of SAHA exposed cultures highlighted that 764 transcripts showed a significant >2.0-fold change at 24 h, which reduced to 36 genes at 14 days. 59% of genes were down-regulated at 24 h and 36% at 14 days, respectively. Pathway analysis indicated SAHA increased expression of members of the matrix metalloproteinase (MMP) family. Furthermore, SAHA-supplementation increased MMP-13 protein expression (7 d, 14 days) and enzyme activity (48 h, 14 days). Selective MMP-13-inhibition (MMP-13i) dose-dependently accelerated mineralization in both SAHA-treated and non-treated cultures. MMP-13i-supplementation promoted expression of several mineralization-associated markers, however, HDACi-induced cell migration and wound healing were impaired. Data demonstrate that short-term low-dose SAHA-exposure promotes mineralization in DPCs by modulating gene pathways and tissue proteases. MMP-13i further increased mineralization-associated events, but decreased HDACi cell migration indicating a specific role for MMP-13 in pulpal repair processes. Pharmacological inhibition of HDAC and MMP may provide novel insights into pulpal repair processes with significant translational benefit. J. Cell. Physiol. 231: 798-816, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Henry F Duncan
- Division of Restorative Dentistry and Periodontology, Dublin Dental University Hospital, Trinity College Dublin, Lincoln Place, Dublin 2, Ireland
| | - Anthony J Smith
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK
| | - Garry J P Fleming
- Material Science Unit, Dublin Dental University Hospital, Trinity College Dublin, Ireland
| | - Nicola C Partridge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York
| | - Emi Shimizu
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York
| | - Gary P Moran
- Division of Oral Biosciences, Dublin Dental University Hospital, Trinity College Dublin, Ireland
| | - Paul R Cooper
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK
| |
Collapse
|
27
|
Lin Z, Bishop KS, Sutherland H, Marlow G, Murray P, Denny WA, Ferguson LR. A quinazoline-based HDAC inhibitor affects gene expression pathways involved in cholesterol biosynthesis and mevalonate in prostate cancer cells. MOLECULAR BIOSYSTEMS 2016; 12:839-49. [PMID: 26759180 DOI: 10.1039/c5mb00554j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chronic inflammation can lead to the development of cancers and resolution of inflammation is an ongoing challenge. Inflammation can result from dysregulation of the epigenome and a number of compounds that modify the epigenome are in clinical use. In this study the anti-inflammatory and anti-cancer effects of a quinazoline epigenetic-modulator compound were determined in prostate cancer cell lines using a non-hypothesis driven transcriptomics strategy utilising the Affymetrix PrimeView® Human Gene Expression microarray. GATHER and IPA software were used to analyse the data and to provide information on significantly modified biological processes, pathways and networks. A number of genes were differentially expressed in both PC3 and DU145 prostate cancer cell lines. The top canonical pathways that frequently arose across both cell lines at a number of time points included cholesterol biosynthesis and metabolism, and the mevalonate pathway. Targeting of sterol and mevalonate pathways may be a powerful anticancer approach.
Collapse
Affiliation(s)
- Z Lin
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand.
| | - K S Bishop
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand.
| | - H Sutherland
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand.
| | - G Marlow
- Discipline of Nutrition and Dietetics, University of Auckland, New Zealand
| | - P Murray
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand.
| | - W A Denny
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand.
| | - L R Ferguson
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand. and Discipline of Nutrition and Dietetics, University of Auckland, New Zealand
| |
Collapse
|
28
|
Venkateswaran K, Shrivastava A, Agrawala PK, Prasad A, Kalra N, Pandey PR, Manda K, Raj HG, Parmar VS, Dwarakanath BS. Mitigation of radiation-induced hematopoietic injury by the polyphenolic acetate 7, 8-diacetoxy-4-methylthiocoumarin in mice. Sci Rep 2016; 6:37305. [PMID: 27849061 PMCID: PMC5110976 DOI: 10.1038/srep37305] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/27/2016] [Indexed: 11/09/2022] Open
Abstract
Protection of the hematopoietic system from radiation damage, and/or mitigation of hematopoietic injury are the two major strategies for developing medical countermeasure agents (MCM) to combat radiation-induced lethality. In the present study, we investigated the potential of 7, 8-diacetoxy-4-methylthiocoumarin (DAMTC) to ameliorate radiation-induced hematopoietic damage and the associated mortality following total body irradiation (TBI) in C57BL/6 mice. Administration of DAMTC 24 hours post TBI alleviated TBI-induced myelo-suppression and pancytopenia, by augmenting lymphocytes and WBCs in the peripheral blood of mice, while bone marrow (BM) cellularity was restored through enhanced proliferation of the stem cells. It stimulated multi-lineage expansion and differentiation of myeloid progenitors in the BM and induced proliferation of splenic progenitors thereby, facilitating hematopoietic re-population. DAMTC reduced the radiation-induced apoptotic and mitotic death in the hematopoietic compartment. Recruitment of pro-inflammatory M1 macrophages in spleen contributed to the immune-protection linked to the mitigation of hematopoietic injury. Recovery of the hematopoietic compartment correlated well with mitigation of mortality at a lethal dose of 9 Gy, leading to 80% animal survival. Present study establishes the potential of DAMTC to mitigate radiation-induced injury to the hematopoietic system by stimulating the re-population of stem cells from multiple lineages.
Collapse
Affiliation(s)
- Kavya Venkateswaran
- Division of Metabolic Cell Signalling Research, Institute of Nuclear Medicine and Allied Sciences, Brig. S. K. Mazumdar Marg, Lucknow Road, Delhi 110054, India.,Department of Zoology, University of Delhi, Delhi 110007, India
| | | | - Paban K Agrawala
- Division of Metabolic Cell Signalling Research, Institute of Nuclear Medicine and Allied Sciences, Brig. S. K. Mazumdar Marg, Lucknow Road, Delhi 110054, India
| | - Ashok Prasad
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Namita Kalra
- Division of Metabolic Cell Signalling Research, Institute of Nuclear Medicine and Allied Sciences, Brig. S. K. Mazumdar Marg, Lucknow Road, Delhi 110054, India
| | - Parvat R Pandey
- Division of Metabolic Cell Signalling Research, Institute of Nuclear Medicine and Allied Sciences, Brig. S. K. Mazumdar Marg, Lucknow Road, Delhi 110054, India
| | - Kailash Manda
- Division of Metabolic Cell Signalling Research, Institute of Nuclear Medicine and Allied Sciences, Brig. S. K. Mazumdar Marg, Lucknow Road, Delhi 110054, India
| | - Hanumantharao G Raj
- Department of Biochemistry, VP Chest Institute, University of Delhi, Delhi 110007, India
| | - Virinder S Parmar
- Bioorganic Laboratory, Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Bilikere S Dwarakanath
- Division of Metabolic Cell Signalling Research, Institute of Nuclear Medicine and Allied Sciences, Brig. S. K. Mazumdar Marg, Lucknow Road, Delhi 110054, India.,Central Research Facility, Sri Ramachandra University, Porur, Chennai 600116, India
| |
Collapse
|
29
|
Histone deacetylase inhibitors induce leukemia gene expression in cord blood hematopoietic stem cells expanded ex vivo. Int J Hematol 2016; 105:37-43. [DOI: 10.1007/s12185-016-2075-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 01/06/2023]
|
30
|
Abstract
Evidence presented over the last few years indicates that the hematopoietic stem cell (HSC) compartment comprises not just one but a number of different cell populations. Based on HSCs’ proliferation and engraftment potential, it has been suggested that there are two classes of HSC, with long- and short-term engraftment potential. HSC heterogeneity seems to involve differentiation capacities as well, since it has been shown that some HSC clones are able to give rise to both myeloid and lymphoid progeny, whereas others are lymphoid deficient. It has been recognized that HSC function depends on intrinsic cell regulators, which are modulated by external signals. Among the former, we can include transcription factors and non-coding RNAs as well as epigenetic modifiers. Among the latter, cytokines and extracellular matrix molecules have been implicated. Understanding the elements and mechanisms that regulate HSC populations is of significant relevance both in biological and in clinical terms, and research in this area still has to face several complex and exciting challenges.
Collapse
Affiliation(s)
- Hector Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, IMSS National Medical Center, Mexico City, Mexico
| |
Collapse
|
31
|
Psatha N, Karponi G, Yannaki E. Optimizing autologous cell grafts to improve stem cell gene therapy. Exp Hematol 2016; 44:528-39. [PMID: 27106799 DOI: 10.1016/j.exphem.2016.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 10/21/2022]
Abstract
Over the past decade, stem cell gene therapy has achieved unprecedented curative outcomes for several genetic disorders. Despite the unequivocal success, clinical gene therapy still faces challenges. Genetically engineered hematopoietic stem cells are particularly vulnerable to attenuation of their repopulating capacity once exposed to culture conditions, ultimately leading to low engraftment levels posttransplant. This becomes of particular importance when transduction rates are low or/and competitive transplant conditions are generated by reduced-intensity conditioning in the absence of a selective advantage of the transduced over the unmodified cells. These limitations could partially be overcome by introducing megadoses of genetically modified CD34(+) cells into conditioned patients or by transplanting hematopoietic stem cells hematopoietic stem cells with high engrafting and repopulating potential. On the basis of the lessons gained from cord blood transplantation, we summarize the most promising approaches to date of increasing either the numbers of hematopoietic stem cells for transplantation or/and their engraftability, as a platform toward the optimization of engineered stem cell grafts.
Collapse
Affiliation(s)
- Nikoletta Psatha
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, WA
| | - Garyfalia Karponi
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Medicine, University of Washington, Seattle, WA.
| |
Collapse
|
32
|
Porada CD, Atala AJ, Almeida-Porada G. The hematopoietic system in the context of regenerative medicine. Methods 2015; 99:44-61. [PMID: 26319943 DOI: 10.1016/j.ymeth.2015.08.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/06/2015] [Accepted: 08/23/2015] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSC) represent the prototype stem cell within the body. Since their discovery, HSC have been the focus of intensive research, and have proven invaluable clinically to restore hematopoiesis following inadvertent radiation exposure and following radio/chemotherapy to eliminate hematologic tumors. While they were originally discovered in the bone marrow, HSC can also be isolated from umbilical cord blood and can be "mobilized" peripheral blood, making them readily available in relatively large quantities. While their ability to repopulate the entire hematopoietic system would already guarantee HSC a valuable place in regenerative medicine, the finding that hematopoietic chimerism can induce immunological tolerance to solid organs and correct autoimmune diseases has dramatically broadened their clinical utility. The demonstration that these cells, through a variety of mechanisms, can also promote repair/regeneration of non-hematopoietic tissues as diverse as liver, heart, and brain has further increased their clinical value. The goal of this review is to provide the reader with a brief glimpse into the remarkable potential HSC possess, and to highlight their tremendous value as therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| | - Anthony J Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| |
Collapse
|
33
|
Xie J, Zhang C. Ex vivo expansion of hematopoietic stem cells. SCIENCE CHINA-LIFE SCIENCES 2015; 58:839-53. [PMID: 26246379 DOI: 10.1007/s11427-015-4895-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023]
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) would benefit clinical applications in several aspects, to improve patient survival, utilize cord blood stem cells for adult applications, and selectively propagate stem cell populations after genetic manipulation. In this review we summarize and discuss recent advances in the culture systems of mouse and human HSCs, which include stroma/HSC co-culture, continuous perfusion and fed-batch cultures, and those supplemented with extrinsic ligands, membrane transportable transcription factors, complement components, protein modification enzymes, metabolites, or small molecule chemicals. Some of the expansion systems have been tested in clinical trials. The optimal condition for ex vivo expansion of the primitive and functional human HSCs is still under development. An improved understanding of the mechanisms for HSC cell fate determination and the HSC culture characteristics will guide development of new strategies to overcome difficulties. In the future, development of a combination treatment regimen with agents that enhance self-renewal, block differentiation, and improve homing will be critical. Methods to enhance yields and lower cost during collection and processing should be employed. The employment of an efficient system for ex vivo expansion of HSCs will facilitate the further development of novel strategies for cell and gene therapies including genome editing.
Collapse
Affiliation(s)
- JingJing Xie
- Taishan Scholar Immunology Program, Binzhou Medical University, Yantai, 264003, China
- Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA
| | - ChengCheng Zhang
- Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA.
| |
Collapse
|
34
|
Varagnolo L, Lin Q, Obier N, Plass C, Dietl J, Zenke M, Claus R, Müller AM. PRC2 inhibition counteracts the culture-associated loss of engraftment potential of human cord blood-derived hematopoietic stem and progenitor cells. Sci Rep 2015. [PMID: 26198814 PMCID: PMC4510577 DOI: 10.1038/srep12319] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cord blood hematopoietic stem cells (CB-HSCs) are an outstanding source for transplantation approaches. However, the amount of cells per donor is limited and culture expansion of CB-HSCs is accompanied by a loss of engraftment potential. In order to analyze the molecular mechanisms leading to this impaired potential we profiled global and local epigenotypes during the expansion of human CB hematopoietic stem and progenitor cells (HPSCs). Human CB-derived CD34+ cells were cultured in serum-free medium together with SCF, TPO, FGF, with or without Igfbp2 and Angptl5 (STF/STFIA cocktails). As compared to the STF cocktail, the STFIA cocktail maintains in vivo repopulation capacity of cultured CD34+ cells. Upon expansion, CD34+ cells genome-wide remodel their epigenotype and depending on the cytokine cocktail, cells show different H3K4me3 and H3K27me3 levels. Expanding cells without Igfbp2 and Angptl5 leads to higher global H3K27me3 levels. ChIPseq analyses reveal a cytokine cocktail-dependent redistribution of H3K27me3 profiles. Inhibition of the PRC2 component EZH2 counteracts the culture-associated loss of NOD scid gamma (NSG) engraftment potential. Collectively, our data reveal chromatin dynamics that underlie the culture-associated loss of engraftment potential. We identify PRC2 component EZH2 as being involved in the loss of engraftment potential during the in vitro expansion of HPSCs.
Collapse
Affiliation(s)
- Linda Varagnolo
- Institute of Medical Radiology and Cell Research (MSZ) in the Center for Experimental Molecular Medicine (ZEMM), University of Würzburg, Würzburg, Germany
| | - Qiong Lin
- Department of Cell Biology, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Nadine Obier
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Christoph Plass
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Johannes Dietl
- Department of Gynecology and Obstetrics, Medical University of Würzburg, Germany
| | - Martin Zenke
- Department of Cell Biology, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Rainer Claus
- 1] Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany [2] Department of Medicine, Div. Hematology, Oncology and Stem Cell Transplantation, University of Freiburg Medical Center, Freiburg, Germany
| | - Albrecht M Müller
- Institute of Medical Radiology and Cell Research (MSZ) in the Center for Experimental Molecular Medicine (ZEMM), University of Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Epigenetic regulatory networks determine the fate of dividing hematopoietic stem cells (HSCs). Prior attempts at the ex-vivo expansion of transplantable human HSCs have led to the depletion or at best maintenance of the numbers of HSCs because of the epigenetic events that silence the HSC gene-expression pattern. The purpose of this review is to outline the recent efforts to use small molecules to reprogram cultured CD34 cells so as to expand their numbers. RECENT FINDINGS Chromatin-modifying agents (CMAs) reactivate the gene-expression patterns of HSCs that have been silenced as they divide ex vivo. Increasing evidence indicates that CMAs act not only by promoting HSC symmetrical self-renewal divisions, but also by reprogramming progenitor cells, resulting in greater numbers of HSCs. The use of such CMAs for these purposes has not resulted in malignant transformation of the ex-vivo treated cell product. SUMMARY The silencing of the gene-expression program that determines HSC function after ex-vivo culture can be reversed by reprogramming the progeny of dividing HSCs with transient exposure to CMAs. The successful implementation of this approach provides a strategy which might lead to the development of a clinically relevant means of manufacturing increased numbers of HSCs.
Collapse
Affiliation(s)
- Camelia Iancu-Rubin
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
36
|
Gajzer D, Ross J, Winder L, Navada S, Zhang W, Silverman L, Chaurasia P. Epigenetic and molecular signatures of cord blood CD34+cells treated with histone deacetylase inhibitors. Vox Sang 2015; 110:79-89. [DOI: 10.1111/vox.12303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/01/2015] [Accepted: 04/27/2015] [Indexed: 12/23/2022]
Affiliation(s)
- D. Gajzer
- Division of Hematology/Medical Oncology; Department of Medicine; Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - J. Ross
- Division of Hematology/Medical Oncology; Department of Medicine; Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - L. Winder
- Division of Hematology/Medical Oncology; Department of Medicine; Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - S. Navada
- Division of Hematology/Medical Oncology; Department of Medicine; Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - W. Zhang
- Department of Medicine Bioinformatics Core; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - L. Silverman
- Division of Hematology/Medical Oncology; Department of Medicine; Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - P. Chaurasia
- Division of Hematology/Medical Oncology; Department of Medicine; Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai; New York NY USA
| |
Collapse
|
37
|
Duncan HF, Smith AJ, Fleming GJP, Cooper PR. Epigenetic modulation of dental pulp stem cells: implications for regenerative endodontics. Int Endod J 2015; 49:431-46. [PMID: 26011759 DOI: 10.1111/iej.12475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 05/24/2015] [Indexed: 12/28/2022]
Abstract
Dental pulp stem cells (DPSCs) offer significant potential for use in regenerative endodontics, and therefore, identifying cellular regulators that control stem cell fate is critical to devising novel treatment strategies. Stem cell lineage commitment and differentiation are regulated by an intricate range of host and environmental factors of which epigenetic influence is considered vital. Epigenetic modification of DNA and DNA-associated histone proteins has been demonstrated to control cell phenotype and regulate the renewal and pluripotency of stem cell populations. The activities of the nuclear enzymes, histone deacetylases, are increasingly being recognized as potential targets for pharmacologically inducing stem cell differentiation and dedifferentiation. Depending on cell maturity and niche in vitro, low concentration histone deacetylase inhibitor (HDACi) application can promote dedifferentiation of several post-natal and mouse embryonic stem cell populations and conversely increase differentiation and accelerate mineralization in DPSC populations, whilst animal studies have shown an HDACi-induced increase in stem cell marker expression during organ regeneration. Notably, both HDAC and DNA methyltransferase inhibitors have also been demonstrated to dramatically increase the reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) for use in regenerative therapeutic procedures. As the regulation of cell fate will likely remain the subject of intense future research activity, this review aims to describe the current knowledge relating to stem cell epigenetic modification, focusing on the role of HDACi on alteration of DPSC phenotype, whilst presenting the potential for therapeutic application as part of regenerative endodontic regimens.
Collapse
Affiliation(s)
- H F Duncan
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College, Dublin, Ireland
| | - A J Smith
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK
| | - G J P Fleming
- Material Science Unit, Dublin Dental University Hospital, Trinity College, Dublin, Ireland
| | - P R Cooper
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham, UK
| |
Collapse
|
38
|
Abstract
Historically, platelet transfusion has proven a reliable way to treat patients suffering from thrombocytopenia or similar ailments. An undersupply of donors, however, has demanded alternative platelet sources. Scientists have therefore sought to recapitulate the biological events that convert hematopoietic stem cells into platelets in the laboratory. Such platelets have shown good function and potential for treatment. Yet the number manufactured ex vivo falls well short of clinical application. Part of the reason is the remarkable gaps in our understanding of the molecular mechanisms driving platelet formation. Using several stem cell sources, scientists have progressively clarified the chemical signaling and physical microenvironment that optimize ex vivo platelets and reconstituted them in synthetic environments. Key advances in cell reprogramming and the ability to propagate self-renewal have extended the lifetime of megakaryocytes to increase the pool of platelet progenitors.
Collapse
Affiliation(s)
- P Karagiannis
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - K Eto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
39
|
Saraf S, Araki H, Petro B, Park Y, Taioli S, Yoshinaga KG, Koca E, Rondelli D, Mahmud N. Ex vivo expansion of human mobilized peripheral blood stem cells using epigenetic modifiers. Transfusion 2014; 55:864-74. [PMID: 25363624 DOI: 10.1111/trf.12904] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Epigenetic modifications likely control the fate of hematopoietic stem cells (HSCs). The chromatin-modifying agents (CMAs), 5-aza-2'-deoxycytidine (5azaD) and trichostatin A (TSA), have previously been shown to expand HSCs from cord blood and marrow. Here we assessed whether CMA can also expand HSCs present in growth factor-mobilized human peripheral blood (MPB). STUDY DESIGN AND METHODS 5azaD and TSA were sequentially added to CD34+ MPB cells in the presence of cytokines, and the cells were cultured for 9 days. RESULTS After culture, a 3.6 ± 0.5-fold expansion of CD34+CD90+ cells, a 10.1 ± 0.5-fold expansion of primitive colony-forming unit (CFU)-mix, and a 2.2 ± 0.5-fold expansion of long-term cobblestone-area-forming cells (CAFCs) was observed in 5azaD/TSA-expanded cells. By contrast, cells cultured in cytokines without 5azaD/TSA displayed no expansion; rather, a reduction in CD34+CD90+ cells (0.7 ± 0.1-fold) and CAFCs (0.3 ± 0.1-fold) from their initial numbers was observed. Global hypomethylation corresponding with increased transcript levels of several genes implicated in HSC self-renewal, including HOXB4, GATA2, and EZH2, was observed in 5azaD/TSA-expanded MPB cells in contrast to controls. 5azaD/TSA-expanded MPB cells retained in vivo hematopoietic engraftment capacity. CONCLUSION MPB CD34+ cells from donors can be expanded using 5azaD/TSA, and these expanded cells retain in vivo hematopoietic reconstitution capacity. This strategy may prove to be potentially useful to augment HSC numbers for patients who fail to mobilize.
Collapse
Affiliation(s)
- Santosh Saraf
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,University of Illinois Cancer Center, Chicago, Illinois
| | - Hiroto Araki
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Benjamin Petro
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Youngmin Park
- UI Hospital Stem Cell Laboratory, Blood & Marrow Transplant Program, Chicago, Illinois
| | - Simona Taioli
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Kazumi G Yoshinaga
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Emre Koca
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Damiano Rondelli
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,UI Hospital Stem Cell Laboratory, Blood & Marrow Transplant Program, Chicago, Illinois.,University of Illinois Cancer Center, Chicago, Illinois
| | - Nadim Mahmud
- Section of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.,UI Hospital Stem Cell Laboratory, Blood & Marrow Transplant Program, Chicago, Illinois.,University of Illinois Cancer Center, Chicago, Illinois
| |
Collapse
|