1
|
Li C, Shin H, Bhavanasi D, Liu M, Yu X, Peslak SA, Liu X, Alvarez-Dominguez JR, Blobel GA, Gregory BD, Huang J, Klein PS. Expansion of human hematopoietic stem cells by inhibiting translation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.28.568925. [PMID: 38077058 PMCID: PMC10705409 DOI: 10.1101/2023.11.28.568925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Hematopoietic stem cell (HSC) transplantation using umbilical cord blood (UCB) is a potentially life-saving treatment for leukemia and bone marrow failure but is limited by the low number of HSCs in UCB. The loss of HSCs after ex vivo manipulation is also a major obstacle to gene editing for inherited blood disorders. HSCs require a low rate of translation to maintain their capacity for self-renewal, but hematopoietic cytokines used to expand HSCs stimulate protein synthesis and impair long-term self-renewal. We previously described cytokine-free conditions that maintain but do not expand human and mouse HSCs ex vivo. Here we performed a high throughput screen and identified translation inhibitors that allow ex vivo expansion of human HSCs while minimizing cytokine exposure. Transplantation assays show a ~5-fold expansion of long-term HSCs from UCB after one week of culture in low cytokine conditions. Single cell transcriptomic analysis demonstrates maintenance of HSCs expressing mediators of the unfolded protein stress response, further supporting the importance of regulated proteostasis in HSC maintenance and expansion. This expansion method maintains and expands human HSCs after CRISPR/Cas9 editing of the BCL11A+58 enhancer, overcoming a major obstacle to ex vivo gene correction for human hemoglobinopathies.
Collapse
Affiliation(s)
- Chenchen Li
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanna Shin
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dheeraj Bhavanasi
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mai Liu
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiang Yu
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott A. Peslak
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Xiaolei Liu
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Juan R. Alvarez-Dominguez
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerd A. Blobel
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian D. Gregory
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jian Huang
- Coriell Institute for Medical Research; Camden, NJ, 08103, USA
- Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
| | - Peter S. Klein
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
2
|
Wang Y, Sugimura R. Ex vivo expansion of hematopoietic stem cells. Exp Cell Res 2023; 427:113599. [PMID: 37061173 DOI: 10.1016/j.yexcr.2023.113599] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/27/2023] [Accepted: 04/09/2023] [Indexed: 04/17/2023]
Abstract
Hematopoietic stem cells (HSCs) are multipotent progenitor cells that can differentiate into various mature blood cells and immune cells, thus reconstituting hematopoiesis. By taking advantage of the tremendous potential of HSCs, varied hereditary and hematologic diseases are promised to be alleviated or cured. To solve the contradiction between the growing demand for HSCs in disease treatment and the low population of HSCs in both cord blood and bone marrow, ex vivo HSC expansion along with multiple protocols has been investigated for harvesting adequate HSCs over the past two decades. This review surveys the state-of-the-art techniques for ex vivo HSC self-renewal and provides a concise summary of the effects of diverse intrinsic and extrinsic factors on the expansion of HSCs. The remaining challenges and emerging opportunities in the field of HSC expansion are also presented.
Collapse
Affiliation(s)
- Yuan Wang
- Centre for Translational Stem Cell Biology, Hong Kong
| | - Ryohichi Sugimura
- Centre for Translational Stem Cell Biology, Hong Kong; Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong.
| |
Collapse
|
3
|
Effects of glucose on the proliferation of human umbilical cord blood hematopoietic stem cells. Cell Tissue Bank 2022; 24:485-494. [DOI: 10.1007/s10561-022-10048-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022]
|
4
|
Yang J, Shen G, Cao J, Zhang J, Gu Y, Zhang X, Jiang X, Luo M, Lu Z. Efficient expansion of mouse hematopoietic stem cells ex vivo by membrane anchored Angptl2. Biochem Biophys Res Commun 2022; 617:42-47. [DOI: 10.1016/j.bbrc.2022.05.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022]
|
5
|
Xuan J, Liu Y, Liu J, Zeng X, Wang H. New Insights into Hematopoietic Stem Cell Expansion to Stimulate Repopulation of the Adult Blood System for Transplantation. Life (Basel) 2022; 12:life12050716. [PMID: 35629383 PMCID: PMC9146250 DOI: 10.3390/life12050716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Successful engraftment of hematopoietic stem cells (HSCs) and progenitor cells (HSPCs) may be considered as a basis for the repopulation of the blood cells after transplantation in adults. Therefore, in vivo and ex vivo expansion of HSCs holds great promise for clinical applications. In this review, the mechanisms of HSC expansion will be discussed, considering the previous studies and works of literature. This is aimed to identify the signaling pathways that regulate HSC expansion and improve the application of engraftment in disease management. The following aspects will be included: (i) Stimulation of HSCs growth in vivo through gene regulation and cytokines activation; (ii) direct or indirect induction of HSC expansion by regulating signaling pathways; (iii) addition to assisting cells to help in the proliferation of HSCs; (iv) changing of living environment in the HSCs cultures via adjusting components and forms of cultures; (v) enhancement of HSC expansion by incorporating substances, such as extracellular vesicles (EVs), UM171, among others. In this review, recent new findings that provide us with new insights into HSC expansion methods have been summarized. Furthermore, these findings will also provide more possibilities for the development of some novel strategies for expanding and engrafting HSCs applied for treatments of some hematopoietic disorders.
Collapse
Affiliation(s)
- Jiangying Xuan
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Yingxia Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
| | - Jinhui Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
| | - Xiaoping Zeng
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
| | - Hongmei Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
- Correspondence: ; Tel.: +86-137-6700-4966
| |
Collapse
|
6
|
Albayrak E, Uslu M, Akgol S, Tuysuz EC, Kocabas F. Small molecule-mediated modulation of ubiquitination and neddylation improves HSC function ex vivo. J Cell Physiol 2021; 236:8122-8136. [PMID: 34101829 DOI: 10.1002/jcp.30466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 11/07/2022]
Abstract
Hematopoietic stem cells (HSCs) are particularly characterized by their quiescence and self-renewal. Cell cycle regulators tightly control quiescence and self-renewal capacity. Studies suggest that modulation of ubiquitination and neddylation could contribute to HSC function via cyclin-dependent kinase inhibitors (CDKIs). S-phase kinase-associated protein 2 (SKP2) is responsible for ubiquitin-mediated proteolysis of CDKIs. Here, we modulated overall neddylation and SKP2-associated ubiquitination in HSCs by using SKP2-C25, an SKP2 inhibitor, and MLN4924 (Pevonedistat) as an inhibitor of the NEDD8 system. Treatments of SKP2-C25 and MLN4924 increased both murine and human stem and progenitor cell (HSPC) compartments. This is associated with the improved quiescence of murine HSC by upregulation of p27 and p57 CDKIs. A colony-forming unit assay showed an enhanced in vitro self-renewal potential post inhibition of ubiquitination and neddylation. In addition, MLN4924 triggered the mobilization of bone marrow HSPCs to peripheral blood. Intriguingly, MLN4924 treatment could decrease the proliferation of murine bone marrow mesenchymal stem cells or endothelial cells. These findings shed light on the contribution of SKP2, and associated ubiquitination and neddylation in HSC maintenance, self-renewal, and expansion.
Collapse
Affiliation(s)
- Esra Albayrak
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Merve Uslu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Sezer Akgol
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Emre Can Tuysuz
- Department of Medical Genetics, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Fatih Kocabas
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
7
|
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: 3.7] [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
|
8
|
Udalamaththa VL, Kaluarachchi A, Wijeratne S, Udagama PV. Therapeutic uses of post-partum tissue-derived mesenchymal stromal cell secretome. Indian J Med Res 2020; 152:541-552. [PMID: 34145093 PMCID: PMC8224162 DOI: 10.4103/ijmr.ijmr_1450_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Indexed: 12/24/2022] Open
Abstract
Human post-partum tissue mesenchymal stromal cells (hPPT-MSCs) are widely used in research to investigate their differentiation capabilities and therapeutic effects as potential agents in cell-based therapy. This is ascribed to the advantages offered by the use of MSCs isolated from hPPT over other MSC sources. A paradigm shift in related research is evident that focuses on the secretome of the human MSCs (hMSCs), as therapeutic effects of hMSCs are attributed more so to their secreted growth factors, cytokines and chemokines and to the extracellular vesicles (EVs), all of which are components of the hMSC secretome. Positive therapeutic effects of the hPPT-MSC secretome have been demonstrated in diseases related to skin, kidney, heart, nervous system, cartilage and bones, that have aided fast recovery by replacing damaged, non-functional tissues, via differentiating and regenerating cells. Although certain limitations such as short half -life of the secretome components and irregular secreting patterns exist in secretome therapy, these issues are successfully addressed with the use of cutting-edge technologies such as genome editing and recombinant cytokine treatment. If the current limitations can be successfully overcome, the hPPT-MSC secretome including its EVs may be developed into a cost-effective therapeutic agent amenable to be used against a wide range of diseases/disorders.
Collapse
Affiliation(s)
| | - Athula Kaluarachchi
- Department of Obstetrics & Gynaecology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | | | - Preethi Vidya Udagama
- Department of Zoology & Environment Sciences, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
9
|
Sun S, Jiang N, Jiang Y, He Q, He H, Wang X, Yang L, Li R, Liu F, Lin X, Zhao B. Chromatin remodeler Znhit1 preserves hematopoietic stem cell quiescence by determining the accessibility of distal enhancers. Leukemia 2020; 34:3348-3358. [PMID: 32694618 PMCID: PMC7685981 DOI: 10.1038/s41375-020-0988-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 01/10/2023]
Abstract
Hematopoietic stem cell (HSC) utilizes its quiescence feature to combat exhaustion for lifetime blood cell supply. To date, how certain chromatin architecture and subsequent transcription profile permit HSC quiescence remains unclear. Here, we show an essential role of chromatin remodeler zinc finger HIT-type containing 1 (Znhit1) in maintaining HSC quiescence. We find that loss of Znhit1 leads to exhaustion of stem cell pool and impairment of hematopoietic function. Mechanically, Znhit1 determines the chromatin accessibility at distal enhancers of HSC quiescence genes, including Pten, Fstl1, and Klf4, for sustained transcription and consequent PI3K-Akt signaling inhibition. Moreover, Znhit1-Pten-PI3K-Akt axis also participates in controlling myeloid expansion and B-lymphoid specification. Our findings therefore identify a dominant role of Znhit1-mediated chromatin remodeling in preserving HSC function for hematopoietic homeostasis.
Collapse
Affiliation(s)
- Shenfei Sun
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.,National Health Commission Key Laboratory of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Shanghai, 200032, China
| | - Ning Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yamei Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Qiuping He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hua He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xin Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Li Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Runsheng Li
- National Health Commission Key Laboratory of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Shanghai, 200032, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
10
|
Pagliuca S, Ruggeri A, Peffault de Latour R. Cord blood transplantation for bone marrow failure syndromes: state of art. Stem Cell Investig 2019; 6:39. [PMID: 32039261 DOI: 10.21037/sci.2019.10.04] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) and immunosuppressive therapy (IST) represent the milestones of the treatment algorithm for idiopathic and inherited bone marrow failure (BMF) disorders. However, patients lacking a suitable donor or failing IST still have a poor prognosis. Cord blood transplantation (CBT) has extended the possibility of HSCT for many patients in case of the absence of an eligible donor, and although in the last years, this procedure is less used in several hematological diseases, it remains an option for the treatment of patients with BMF syndromes. Nevertheless, optimization of conditioning regimen and cord blood unit selection is warranted to reduce the risk of graft failure and transplant-related mortality. This review summarizes the state of art of CBT in the field of BMF diseases, focusing on historical and recent issues in idiopathic aplastic anemia and inherited disorders.
Collapse
Affiliation(s)
- Simona Pagliuca
- Hematology and Transplantation Unit, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Annalisa Ruggeri
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy.,Eurocord-Monacord, Hôpital Saint Louis, Paris, France.,Cellular Therapy and Immunobiology Working Party of EBMT, Leiden, The Netherlands
| | - Régis Peffault de Latour
- Hematology and Transplantation Unit, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
11
|
Zhou L, Che Z, Zhang X, Zhou P, Li X, Xu X, Shi Q, Li D, Ju X. Influence of neonatal gender on cord blood CD34 + cell amplification and gene expression. Exp Ther Med 2019; 18:105-118. [PMID: 31258643 PMCID: PMC6566080 DOI: 10.3892/etm.2019.7549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
The present study attempted to evaluate whether neonatal gender affects the hematopoietic potential of cord blood (CB) transplants and, if so, to determine the underlying molecular mechanisms. CD34+ cells from CB were isolated and divided into male and female groups. CD34+CD38− cell populations were then compared using fluorescence-assisted cell sorting (FACS) and a colony formation assay was performed. Next, a Genechip microarray analysis was used to identify differentially expressed genes (DEGs). Finally, the Genechip results were validated by FACS analysis. It was revealed that the male group had higher amplification efficiency. Gene ontology analysis indicated differences in the biological function of the DEGs between the two groups. Kyoto Encyclopedia of Genes and Genomes analysis suggested that the hematopoietic cell lineage signaling pathway was upregulated in the male group along with high expression levels of genes including interleukin (IL) 6 signal transducer (glycoprotein 130), IL-7 and IL-7 receptor. It was speculated that this may be partially due to numerous upregulated DEGs being involved in chromosomal segregation and hematopoietic cell lineage signaling pathways in CD34+ cells from the male group.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Pediatrics, The Sixth People's Hospital of Ji'nan, Jinan, Shandong 250200, P.R. China.,Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhe Che
- Health and Family Planning Inspection Agency of Zhangqiu District, Jinan, Shandong 250200, P.R. China
| | - Xiaowei Zhang
- Department of Pediatrics, The Sixth People's Hospital of Ji'nan, Jinan, Shandong 250200, P.R. China
| | - Panpan Zhou
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xue Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xuejing Xu
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qing Shi
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Stem Cell and Regenerative Medicine Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiuli Ju
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Stem Cell and Regenerative Medicine Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
12
|
Jalnapurkar S, Moirangthem RD, Singh S, Limaye L, Kale V. Microvesicles Secreted by Nitric Oxide-Primed Mesenchymal Stromal Cells Boost the Engraftment Potential of Hematopoietic Stem Cells. Stem Cells 2018; 37:128-138. [PMID: 30290030 DOI: 10.1002/stem.2912] [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: 06/05/2018] [Revised: 08/04/2018] [Accepted: 08/18/2018] [Indexed: 12/23/2022]
Abstract
Patients with leukemia, lymphoma, severe aplastic anemia, etc. are frequently the targets of bone marrow transplantation, the success of which critically depends on efficient engraftment by transplanted hematopoietic cells (HSCs). Ex vivo manipulation of HSCs to improve their engraftment ability becomes necessary when the number or quality of donor HSCs is a limiting factor. Due to their hematopoiesis-supportive ability, bone marrow-derived mesenchymal stromal cells (MSCs) have been traditionally used as feeder layers for ex vivo expansion of HSCs. MSCs form a special HSC-niche in vivo, implying that signaling mechanisms operative in them would affect HSC fate. We have recently demonstrated that AKT signaling prevailing in the MSCs affect the HSC functionality. Here we show that MSCs primed with nitric oxide donor, Sodium nitroprusside (SNP), significantly boost the engraftment potential of the HSCs co-cultured with them via intercellular transfer of microvesicles (MVs) harboring mRNAs encoding HSC-supportive genes. Our data suggest that these MVs could be used as HSC-priming agents to improve transplantation efficacy. Since both, nitric oxide donors and MSCs are already in clinical use; their application in clinical settings may be relatively straight forward. This approach could also be applied in regenerative medicine protocols. Stem Cells 2019;37:128-138.
Collapse
Affiliation(s)
| | | | - Shweta Singh
- Stem Cell Lab, National Centre for Cell Science, Pune, India
| | - Lalita Limaye
- Stem Cell Lab, National Centre for Cell Science, Pune, India
| | - Vaijayanti Kale
- Stem Cell Lab, National Centre for Cell Science, Pune, India
| |
Collapse
|
13
|
Brazvan B, Ebrahimi-Kalan A, Velaei K, Mehdipour A, Aliyari Serej Z, Ebrahimi A, Ghorbani M, Cheraghi O, Nozad Charoudeh H. Telomerase activity and telomere on stem progeny senescence. Biomed Pharmacother 2018; 102:9-17. [PMID: 29547744 DOI: 10.1016/j.biopha.2018.02.073] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/03/2018] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
The end of linear chromosomes is formed of a special nucleoprotein heterochromatin structure with repetitive TTAGGG sequences called telomere. Telomere length is regulated by a special enzyme called telomerase, a specific DNA polymerase that adds new telomeric sequences to the chromosome ends. Telomerase consists of two parts; the central protein part and the accessory part which is a RNA component transported by the central part. Regulation of telomere length by this enzyme is a multi-stage process. Telomere length elongation is strongly influenced by the level of telomerase and has a strong correlation with the activity of telomerase enzyme. Human Telomerase Reverse Transcriptase (hTERT) gene expression plays an important role in maintaining telomere length and high proliferative property of cells. Except a low activity of telomerase enzyme in hematopoietic and few types of stem cells, most of somatic cells didn't showed telomerase activity. Moreover, cytokines are secretory proteins that control many aspects of hematopoiesis, especially immune responses and inflammation. Also, the induction of hTERT gene expression by cytokines is organized through the PI3K/AKT and NF/kB signaling pathways. In this review we have tried to talk about effects of immune cell cytokines on telomerase expression/telomere length and the induction of telomerase expression by cytokines.
Collapse
Affiliation(s)
- Balal Brazvan
- Department of Basic Sciences, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kobra Velaei
- Department of Anatomical Science, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mehdipour
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeynab Aliyari Serej
- Applied Cell Sciences Department, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayyub Ebrahimi
- Department of Molecular Biology and Genetic, Faculty of Arts and Sciences, Halic Uuniversity, Istanbul, Turkey
| | - Mohammad Ghorbani
- Department of Basic Sciences, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Omid Cheraghi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| | | |
Collapse
|
14
|
Sieber S, Wirth L, Cavak N, Koenigsmark M, Marx U, Lauster R, Rosowski M. Bone marrow-on-a-chip: Long-term culture of human haematopoietic stem cells in a three-dimensional microfluidic environment. J Tissue Eng Regen Med 2017; 12:479-489. [PMID: 28658717 DOI: 10.1002/term.2507] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 12/17/2022]
Abstract
Multipotent haematopoietic stem and progenitor cells (HSPCs) are the source for all blood cell types. The bone marrow stem cell niche in which the HSPCs are maintained is known to be vital for their maintenance. Unfortunately, to date, no in vitro model exists that accurately mimics the aspects of the bone marrow niche and simultaneously allows the long-term culture of HSPCs. In this study, a novel three-dimensional coculture model is presented, based on a hydroxyapatite coated zirconium oxide scaffold, comprising of human mesenchymal stromal cells (MSCs) and cord blood derived HSPCs, enabling successful HSPC culture for a time span of 28 days within the microfluidic multiorgan chip. The HSPCs were found to stay in their primitive state (CD34+ CD38- ) and capable of granulocyte, erythrocyte, macrophage, megakaryocyte colony formation. Furthermore, a microenvironment was formed bearing molecular and structural similarity to the in vivo bone marrow niche containing extracellular matrix and signalling molecules known to play an important role in HSPC homeostasis. Here, a novel human in vitro bone marrow model is presented for the first time, capable of long-term culture of primitive HSPCs in a microfluidic environment.
Collapse
Affiliation(s)
- Stefan Sieber
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charitè Campus Virchow Klinikum, Berlin, Germany
| | - Lorenz Wirth
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Nino Cavak
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Marielle Koenigsmark
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | | | - Roland Lauster
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Mark Rosowski
- Department Medical Biotechnology, Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| |
Collapse
|
15
|
Ratajczak MZ. Why are hematopoietic stem cells so 'sexy'? on a search for developmental explanation. Leukemia 2017; 31:1671-1677. [PMID: 28502982 PMCID: PMC5540746 DOI: 10.1038/leu.2017.148] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/03/2017] [Accepted: 03/07/2017] [Indexed: 02/07/2023]
Abstract
Evidence has accumulated that normal human and murine hematopoietic stem cells express several functional pituitary and gonadal sex hormones, and that, in fact, some sex hormones, such as androgens, have been employed for many years to stimulate hematopoiesis in patients with bone marrow aplasia. Interestingly, sex hormone receptors are also expressed by leukemic cell lines and blasts. In this review, I will discuss the emerging question of why hematopoietic cells express these receptors. A tempting hypothetical explanation for this phenomenon is that hematopoietic stem cells are related to subpopulation of migrating primordial germ cells. To support of this notion, the anatomical sites of origin of primitive and definitive hematopoiesis during embryonic development are tightly connected with the migratory route of primordial germ cells: from the proximal epiblast to the extraembryonic endoderm at the bottom of the yolk sac and then back to the embryo proper via the primitive streak to the aorta-gonado-mesonephros (AGM) region on the way to the genital ridges. The migration of these cells overlaps with the emergence of primitive hematopoiesis in the blood islands at the bottom of the yolk sac, and definitive hematopoiesis that occurs in hemogenic endothelium in the embryonic dorsal aorta in AGM region.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
16
|
Kiernan J, Damien P, Monaghan M, Shorr R, McIntyre L, Fergusson D, Tinmouth A, Allan D. Clinical Studies of Ex Vivo Expansion to Accelerate Engraftment After Umbilical Cord Blood Transplantation: A Systematic Review. Transfus Med Rev 2016; 31:173-182. [PMID: 28087163 DOI: 10.1016/j.tmrv.2016.12.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/30/2016] [Accepted: 12/20/2016] [Indexed: 01/04/2023]
Abstract
Cell dose limits greater use of umbilical cord blood (UCB) in hematopoietic cell transplantation. The clinical benefits of ex vivo expansion need clarity to understand its potential impact. A systematic search of studies addressing UCB ex vivo expansion was conducted. Fifteen clinical studies (349 transplanted patients) and 13 registered trials were identified. The co-infusion of an expanded unit and a second unmanipulated unit (8 studies), the fractional expansion of 12% to 60% of a single unit (5 studies), and the infusion of a single expanded unit (2 studies) were reported. More recently, published studies and 12 of 13 ongoing trials involve the use of novel small molecules in addition to traditional cytokine cocktails. Higher total cell number was closely associated with faster neutrophil engraftment. Compared with historical controls, neutrophil engraftment was significantly accelerated in more recent studies using small molecules or mesenchymal stromal cells (MSC) co-culture, and in some cases, platelet recovery was also statistically improved. Recent studies using nicotinamide and StemRegenin-1 reported long-term chimerism of the expanded unit. No significant improvement in survival or other transplant-related outcomes was demonstrated for any of the strategies. Ex vivo expansion of UCB can accelerate initial neutrophil engraftment after transplant. More recent studies suggest that long-term engraftment of ex vivo expanded cord blood units is achievable. Results of larger randomized controlled trials are needed to understand the impact on patient outcomes and health care costs.
Collapse
Affiliation(s)
- Jeffrey Kiernan
- Center for Transfusion Research, University of Ottawa, Ottawa, Ontario, Canada; The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Pauline Damien
- Center for Transfusion Research, University of Ottawa, Ottawa, Ontario, Canada; The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | - Risa Shorr
- Medical Library Services, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Lauralyn McIntyre
- Center for Transfusion Research, University of Ottawa, Ottawa, Ontario, Canada; The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean Fergusson
- Center for Transfusion Research, University of Ottawa, Ottawa, Ontario, Canada; The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alan Tinmouth
- Center for Transfusion Research, University of Ottawa, Ottawa, Ontario, Canada; The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David Allan
- Center for Transfusion Research, University of Ottawa, Ottawa, Ontario, Canada; The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
17
|
Galeev R, Baudet A, Kumar P, Rundberg Nilsson A, Nilsson B, Soneji S, Törngren T, Borg Å, Kvist A, Larsson J. Genome-wide RNAi Screen Identifies Cohesin Genes as Modifiers of Renewal and Differentiation in Human HSCs. Cell Rep 2016; 14:2988-3000. [PMID: 26997282 DOI: 10.1016/j.celrep.2016.02.082] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/04/2016] [Accepted: 02/22/2016] [Indexed: 12/21/2022] Open
Abstract
To gain insights into the regulatory mechanisms of hematopoietic stem cells (HSCs), we employed a genome-wide RNAi screen in human cord-blood derived cells and identified candidate genes whose knockdown maintained the HSC phenotype during culture. A striking finding was the identification of members of the cohesin complex (STAG2, RAD21, STAG1, and SMC3) among the top 20 genes from the screen. Upon individual validation of these cohesin genes, we found that their knockdown led to an immediate expansion of cells with an HSC phenotype in vitro. A similar expansion was observed in vivo following transplantation to immunodeficient mice. Transcriptome analysis of cohesin-deficient CD34(+) cells showed an upregulation of HSC-specific genes, demonstrating an immediate shift toward a more stem-cell-like gene expression signature upon cohesin deficiency. Our findings implicate cohesin as a major regulator of HSCs and illustrate the power of global RNAi screens to identify modifiers of cell fate.
Collapse
Affiliation(s)
- Roman Galeev
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Aurélie Baudet
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Praveen Kumar
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | | | - Björn Nilsson
- Division of Hematology and Transfusion Medicine, Lund University, 221 84 Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Therese Törngren
- Division of Oncology and Pathology, Lund University, 223 63 Lund, Sweden
| | - Åke Borg
- Division of Oncology and Pathology, Lund University, 223 63 Lund, Sweden
| | - Anders Kvist
- Division of Oncology and Pathology, Lund University, 223 63 Lund, Sweden
| | - Jonas Larsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden.
| |
Collapse
|
18
|
Tiwari A, Tursky ML, Nekkanti LP, Jenkin G, Kirkland MA, Pande G. Expansion of Human Hematopoietic Stem/Progenitor Cells on Decellularized Matrix Scaffolds. ACTA ACUST UNITED AC 2016; 36:1C.15.1-1C.15.16. [PMID: 26840223 DOI: 10.1002/9780470151808.sc01c15s36] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Umbilical cord blood (UCB) is one of the richest sources for hematopoietic stem/progenitor cells (HSPCs), with more than 3000 transplantations performed each year for the treatment of leukemia and other bone marrow, immunological, and hereditary diseases. However, transplantation of single cord blood units is mostly restricted to children, due to the limited number of HSPC per unit. This unit develops a method to increase the number of HSPCs in laboratory conditions by using cell-free matrices from bone marrow cells that mimic 'human-body-niche-like' conditions as biological scaffolds to support the ex vivo expansion of HSPCs. In this unit, we describe protocols for the isolation and characterization of HSPCs from UCB and their serum-free expansion on decellularized matrices. This method may also help to provide understanding of the biochemical organization of hematopoietic niches and lead to suggestions regarding the design of tissue engineering-based biomimetic scaffolds for HSPC expansion for clinical applications.
Collapse
Affiliation(s)
- Abhilasha Tiwari
- CSIR Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India.,Geelong Technology Precinct, Deakin University, Geelong, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, Australia
| | - Melinda L Tursky
- Lowy Cancer Research Centre, University of New South Wales, New South Wales, Australia
| | - Lakshmi P Nekkanti
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, Australia
| | - Mark A Kirkland
- Geelong Technology Precinct, Deakin University, Geelong, Victoria, Australia
| | - Gopal Pande
- CSIR Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India
| |
Collapse
|
19
|
Pelagiadis I, Stiakaki E, Choulaki C, Kalmanti M, Dimitriou H. The role of children's bone marrow mesenchymal stromal cells in the ex vivo expansion of autologous and allogeneic hematopoietic stem cells. Cell Biol Int 2015; 39:1099-110. [DOI: 10.1002/cbin.10483] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/08/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Iordanis Pelagiadis
- Department of Pediatric Hematology-Oncology; Medical School; University of Crete; Heraklion Crete Greece
| | - Eftichia Stiakaki
- Department of Pediatric Hematology-Oncology; Medical School; University of Crete; Heraklion Crete Greece
| | - Christianna Choulaki
- Department of Pediatric Hematology-Oncology; Medical School; University of Crete; Heraklion Crete Greece
| | - Maria Kalmanti
- Department of Pediatric Hematology-Oncology; Medical School; University of Crete; Heraklion Crete Greece
| | - Helen Dimitriou
- Department of Pediatric Hematology-Oncology; Medical School; University of Crete; Heraklion Crete Greece
| |
Collapse
|
20
|
Predictions for optimal mitigation of paracrine inhibitory signalling in haemopoietic stem cell cultures. Stem Cell Res Ther 2015; 6:58. [PMID: 25888759 PMCID: PMC4443622 DOI: 10.1186/s13287-015-0048-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/27/2015] [Accepted: 03/10/2015] [Indexed: 01/01/2023] Open
Abstract
Introduction Recent studies in the literature have highlighted the critical role played by cell signalling in determining haemopoietic stem cell (HSC) fate within ex vivo culture systems. Stimulatory signals can enhance proliferation and promote differentiation, whilst inhibitory signals can significantly limit culture output. Methods Numerical models of various mitigation strategies are presented and applied to determine effectiveness of these strategies toward mitigation of paracrine inhibitory signalling inherent in these culture systems. The strategies assessed include mixing, media-exchange, fed-batch and perfusion. Results The models predict that significant spatial concentration gradients exist in typical cell cultures, with important consequences for subsequent cell expansion. Media exchange is shown to be the most effective mitigation strategy, but remains labour intensive and difficult to scale-up for large culture systems. The fed-batch strategy is only effective at very small Peclet number, and its effect is diminished as the cell culture volume grows. Conversely, mixing is effective at high Peclet number, and ineffective at low Peclet number. The models predict that cell expansion in fed-batch cultures becomes independent of increasing dilution rate, consistent with experimental results previously reported in the literature. In contrast, the models predict that increasing the flow rate in perfused cultures will lead to increased cell expansion, indicating the suitability of perfusion for use as an automated, tunable strategy. The effect of initial cell seeding density is also investigated, with the model showing that perfusion outperforms dilution for all densities considered. Conclusions The models predict that the impact of inhibitory signalling in HSC cultures can be mitigated against using media manipulation strategies, with the optimal strategy dependent upon the protein diffusion time-scale relative to the media manipulation time-scale. The key messages from this study can be applied to any complex cell culture scenario where cell-cell interactions and paracrine signalling networks impact upon cell fate and cell expansion. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0048-7) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Bari S, Seah KKH, Poon Z, Cheung AMS, Fan X, Ong SY, Li S, Koh LP, Hwang WYK. Expansion and homing of umbilical cord blood hematopoietic stem and progenitor cells for clinical transplantation. Biol Blood Marrow Transplant 2014; 21:1008-19. [PMID: 25555449 DOI: 10.1016/j.bbmt.2014.12.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022]
Abstract
The successful expansion of hematopoietic stem and progenitor cells (HSPCs) from umbilical cord blood (UCB) for transplantation could revolutionize clinical practice by improving transplantation-related outcomes and making available UCB units that have suboptimal cell doses for transplantation. New cytokine combinations appear able to promote HSPC growth with minimal differentiation into mature precursors and new agents, such as insulin-like growth factor-binding protein 2, are being used in clinical trials. Molecules that simulate the HSPC niche, such as Notch ligand, have also shown promise. Further improvements have been made with the use of mesenchymal stromal cells, which have made possible UCB expansion without a potentially deleterious prior CD34/CD133 cell selection step. Chemical molecules, such as copper chelators, nicotinamide, and aryl hydrocarbon antagonists, have shown excellent outcomes in clinical studies. The use of bioreactors could further add to HSPC studies in future. Drugs that could improve HSPC homing also appear to have potential in improving engraftment times in UCB transplantation. Technologies to expand HSPC from UCB and to enhance the homing of these cells appear to have attained the goal of accelerating hematopoietic recovery. Further discoveries and clinical studies are likely to make the goal of true HSPC expansion a reality for many applications in future.
Collapse
Affiliation(s)
- Sudipto Bari
- Department of Hematology, Singapore General Hospital, Singapore; Department of Pharmacy, National University of Singapore, Singapore
| | | | - Zhiyong Poon
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research and Technology, Singapore
| | | | - Xiubo Fan
- Department of Clinical Research, Singapore General Hospital, Singapore
| | - Shin-Yeu Ong
- Department of Hematology, Singapore General Hospital, Singapore
| | - Shang Li
- Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore
| | - Liang Piu Koh
- Department of Hematology-Oncology, National University Cancer Institute, Singapore
| | - William Ying Khee Hwang
- Department of Hematology, Singapore General Hospital, Singapore; Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore; Singapore Cord Blood Bank, Singapore.
| |
Collapse
|
22
|
Bird GA, Polsky A, Estes P, Hanlon T, Hamilton H, Morton JJ, Gutman J, Jimeno A, Turner BC, Refaeli Y. Expansion of human and murine hematopoietic stem and progenitor cells ex vivo without genetic modification using MYC and Bcl-2 fusion proteins. PLoS One 2014; 9:e105525. [PMID: 25170611 PMCID: PMC4149411 DOI: 10.1371/journal.pone.0105525] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/23/2014] [Indexed: 12/21/2022] Open
Abstract
The long-term repopulating hematopoietic stem cell (HSC) population can self-renew in vivo, support hematopoiesis for the lifetime of the individual, and is of critical importance in the context of bone marrow stem cell transplantation. The mechanisms that regulate the expansion of HSCs in vivo and in vitro remain unclear to date. Since the current set of surface markers only allow for the identification of a population of cells that is highly enriched for HSC activity, we will refer to the population of cells we expand as Hematopoietic Stem and Progenitor cells (HSPCs). We describe here a novel approach to expand a cytokine-dependent Hematopoietic Stem and Progenitor Cell (HSPC) population ex vivo by culturing primary adult human or murine HSPCs with fusion proteins including the protein transduction domain of the HIV-1 transactivation protein (Tat) and either MYC or Bcl-2. HSPCs obtained from either mouse bone marrow, human cord blood, human G-CSF mobilized peripheral blood, or human bone marrow were expanded an average of 87 fold, 16.6 fold, 13.6 fold, or 10 fold, respectively. The expanded cell populations were able to give rise to different types of colonies in methylcellulose assays in vitro, as well as mature hematopoietic populations in vivo upon transplantation into irradiated mice. Importantly, for both the human and murine case, the ex vivo expanded cells also gave rise to a self-renewing cell population in vivo, following initial transplantation, that was able to support hematopoiesis upon serial transplantation. Our results show that a self-renewing cell population, capable of reconstituting the hematopoietic compartment, expanded ex vivo in the presence of Tat-MYC and Tat-Bcl-2 suggesting that this may be an attractive approach to expand human HSPCs ex vivo for clinical use.
Collapse
Affiliation(s)
- Gregory A. Bird
- Taiga Biotechnologies, Inc., Aurora, Colorado, United States of America
| | - Avital Polsky
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology and Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Patricia Estes
- Taiga Biotechnologies, Inc., Aurora, Colorado, United States of America
| | - Teri Hanlon
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology and Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Haley Hamilton
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology and Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - John J. Morton
- University of Colorado School of Medicine, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, United States of America
| | - Jonathan Gutman
- University of Colorado School of Medicine, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, United States of America
| | - Antonio Jimeno
- University of Colorado School of Medicine, Department of Medicine, Division of Medical Oncology, Aurora, Colorado, United States of America
| | - Brian C. Turner
- Taiga Biotechnologies, Inc., Aurora, Colorado, United States of America
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology and Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Yosef Refaeli
- Taiga Biotechnologies, Inc., Aurora, Colorado, United States of America
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology and Department of Dermatology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
23
|
Abstract
A better understanding of the interaction between extrinsic factors and surface receptors on stem cells will greatly benefit stem cell research and applications. Recently, we showed that several angiopoietin-like proteins (Angptls) bind and activate the immune inhibitory receptor human leukocyte immunoglobulin (Ig)-like receptor B2 (LILRB2) to support ex vivo expansion of hematopoietic stem cells (HSCs) and leukemia development. However, the molecular basis for the interaction between Angptls and LILRB2 was unclear. Here, we demonstrate that Angptl2 expressed in mammalian cells forms high-molecular-weight species and that ligand multimerization is required for activation of LILRB2 for downstream signaling. A novel motif in the first and fourth Ig domains of LILRB2 was identified that is necessary for the receptor to be bound and activated by Angptl2. The binding of Angptl2 to LILRB2 is more potent than and not completely overlapped with the binding of another ligand, HLA-G. Immobilized anti-LILRB2 antibodies induce a more potent activation of LILRB2 than Angptl2, and we developed a serum-free culture containing defined cytokines and immobilized anti-LILRB2 that supports a net expansion of repopulating human cord blood HSCs. Our elucidation of the mode of Angptl binding to LILRB2 enabled the development of a new approach for ex vivo expansion of human HSCs.
Collapse
|
24
|
Abstract
Hematopoietic stem cells (HSCs) have become the most extensively studied stem cells and HSC-based cellular therapy is promising for hematopoietic cancers and hereditary blood disorders. Successful treatment of patients with HSC cells depends on sufficient number of highly purified HSCs and progenitor cells. However, stem cells are a very rare population no matter where they come from. Thus, ex vivo amplification of these HSCs is essential. The heavy demands from more and more patients for HSCs also require industrial-scale expansion of HSCs with lower production cost and higher efficiency. Two main ways to reach that goal: (1) to find clinically applicable, simple and efficient methods (or reagents) to enrich HSCs; (2) to find new developmental regulators and chemical compounds in order to replace the currently used cytokine cocktails for HSCs amplification. In this Editorial review, we would like to introduce the current status of ex vivo expansion of HSCs, particularly focusing on enrichment and culture supplements.
Collapse
|
25
|
Tiwari A, Tursky ML, Kirkland MA, Pande G. Expansion of human hematopoietic stem/progenitor cells on decellularized matrix scaffolds. ACTA ACUST UNITED AC 2014; 28:1C.15.1-1C.15.15. [PMID: 24510768 DOI: 10.1002/9780470151808.sc01c15s28] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Umbilical cord blood (UCB) is one of the richest sources for hematopoietic stem/progenitor cells (HSPCs), with more than 3000 transplantations performed each year for the treatment of leukemia and other bone marrow, immunological, and hereditary diseases. However, transplantation of single cord blood units is mostly restricted to children, due to the limited number of HSPC per unit. This unit develops a method to increase the number of HSPCs in laboratory conditions by using cell-free matrices from bone marrow cells that mimic 'human-body niche-like' conditions as biological scaffolds to support the ex vivo expansion of HSPCs. In this unit, we describe protocols for the isolation and characterization of HSPCs from UCB and their serum-free expansion on decellularized matrices. This method may also help to provide understanding of the biochemical organization of hematopoietic niches and lead to suggestions regarding the design of tissue engineering-based biomimetic scaffolds for HSPC expansion for clinical applications.
Collapse
Affiliation(s)
- Abhilasha Tiwari
- CSIR Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India; Geelong Technology Precinct, Deakin University, Geelong, Victoria, Australia; Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
26
|
Diogo MM, da Silva CL, Cabral JMS. Separation Technologies for Stem Cell Bioprocessing. CELL ENGINEERING 2014. [DOI: 10.1007/978-94-007-7196-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Combination of a haploidentical SCT with an unrelated cord blood unit: a single-arm prospective study. Bone Marrow Transplant 2013; 49:206-11. [DOI: 10.1038/bmt.2013.154] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/13/2013] [Accepted: 03/19/2013] [Indexed: 12/13/2022]
|
28
|
Pinho S, Lacombe J, Hanoun M, Mizoguchi T, Bruns I, Kunisaki Y, Frenette PS. PDGFRα and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. ACTA ACUST UNITED AC 2013; 210:1351-67. [PMID: 23776077 PMCID: PMC3698522 DOI: 10.1084/jem.20122252] [Citation(s) in RCA: 385] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A subset of human Nestin+ mesenchymal stem cells expresses PDGFRα and CD51, and these markers can be used for prospective isolation of these cells. The intermediate filament protein Nestin labels populations of stem/progenitor cells, including self-renewing mesenchymal stem cells (MSCs), a major constituent of the hematopoietic stem cell (HSC) niche. However, the intracellular location of Nestin prevents its use for prospective live cell isolation. Hence it is important to find surface markers specific for Nestin+ cells. In this study, we show that the expression of PDGFRα and CD51 among CD45− Ter119− CD31− mouse bone marrow (BM) stromal cells characterizes a large fraction of Nestin+ cells, containing most fibroblastic CFUs, mesenspheres, and self-renewal capacity after transplantation. The PDGFRα+ CD51+ subset of Nestin+ cells is also enriched in major HSC maintenance genes, supporting the notion that niche activity co-segregates with MSC activity. Furthermore, we show that PDGFRα+ CD51+ cells in the human fetal BM represent a small subset of CD146+ cells expressing Nestin and enriched for MSC and HSC niche activities. Importantly, cultured human PDGFRα+ CD51+ nonadherent mesenspheres can significantly expand multipotent hematopoietic progenitors able to engraft immunodeficient mice. These results thus indicate that the HSC niche is conserved between the murine and human species and suggest that highly purified nonadherent cultures of niche cells may represent a useful novel technology to culture human hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Sandra Pinho
- Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
de Lichtervelde L, Boitano AE, Wang Y, Krastel P, Petersen F, Cooke MP, Schultz PG. Eupalinilide E inhibits erythropoiesis and promotes the expansion of hematopoietic progenitor cells. ACS Chem Biol 2013; 8:866-70. [PMID: 23441826 DOI: 10.1021/cb4000234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Hematopoietic stem cells (HSCs) are the progenitor cells that give rise to all blood cells. The ability to control HSC differentiation has the potential to improve the success of bone marrow transplants and the production of functional blood cells ex vivo. Here we performed an unbiased screen using primary human CD34(+) hematopoietic stem and progenitor cells (HSPCs) to identify natural products that selectively control their differentiation. We identified a plant-derived natural product, eupalinilide E, that promotes the ex vivo expansion of HSPCs and hinders the in vitro development of erythrocytes. This activity was additive with aryl hydrocarbon receptor (AhR) antagonists, which are also known to expand HSCs and currently in clinical development. These findings reveal a new activity for eupalinilide E, and suggest that it may be a useful tool to probe the mechanisms of hematopoiesis and improve the ex vivo production of progenitors for therapeutic purposes.
Collapse
Affiliation(s)
- Lorenzo de Lichtervelde
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines
Road, La Jolla, California 92037, United States
| | - Anthony E. Boitano
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay
Hopkins Drive, San Diego, California 92121, United States
| | - Ying Wang
- Natural Products Unit, Novartis Institutes for Biomedical Research, Novartis
Pharma AG, Switzerland
| | - Philipp Krastel
- Natural Products Unit, Novartis Institutes for Biomedical Research, Novartis
Pharma AG, Switzerland
| | - Frank Petersen
- Natural Products Unit, Novartis Institutes for Biomedical Research, Novartis
Pharma AG, Switzerland
| | - Michael P. Cooke
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay
Hopkins Drive, San Diego, California 92121, United States
| | - Peter G. Schultz
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines
Road, La Jolla, California 92037, United States
| |
Collapse
|
30
|
Yu X, Gu Z, Wang Y, Wang H. New strategies in cord blood cells transplantation. Cell Biol Int 2013; 37:865-74. [PMID: 23589409 DOI: 10.1002/cbin.10114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/22/2013] [Indexed: 12/26/2022]
Abstract
For patients lacking a human leucocyte antigen-matched donor, umbilical cord blood (UCB) is an ideal, alternative source of haematopoietic stem cells (HSCs) for transplantation purposes. UCB has many advantages over bone marrow or peripheral blood taken from volunteer donors. UCB is also an important source of other stem cells, including endothelial progenitors, mesenchymal stem cells, very small embryonic/epiblast-like (VSEL) stem cells, and unrestricted somatic stem cells, which are potentially suitable for regenerative medicine. However, a significant clinical problem is that the number of HSCs in one cord-blood unit is not enough for an adult transplantation. The development of new approaches including use of multiple donors, ex vivo expansion, increasing efficiency of homing and engraftment, retrieving more cells from the placenta and cord blood is of crucial importance for the delayed engraftment after UCB transplantation. In the future, UCB will emerge as a source of cells for cellular therapies associated with tissue repair and regeneration.
Collapse
Affiliation(s)
- Xin Yu
- Blood Transfusion Research Institute, Wuxi Red Cross Blood Center, 109 Xinmin Road, Wuxi, 214021, China
| | | | | | | |
Collapse
|
31
|
Andrade PZ, dos Santos F, Cabral JMS, da Silva CL. Stem cell bioengineering strategies to widen the therapeutic applications of haematopoietic stem/progenitor cells from umbilical cord blood. J Tissue Eng Regen Med 2013; 9:988-1003. [PMID: 23564692 DOI: 10.1002/term.1741] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 01/18/2013] [Accepted: 02/05/2013] [Indexed: 12/11/2022]
Abstract
Umbilical cord blood (UCB) transplantation has observed a significant increase in recent years, due to the unique features of UCB haematopoietic stem/progenitor cells (HSCs) for the treatment of blood-related disorders. However, the low cell numbers available per UCB unit significantly impairs the widespread use of this source for transplantation of adult patients, resulting in graft failure, delayed engraftment and delayed immune reconstitution. In order to overcome this issue, distinct approaches are now being considered in clinical trials, such as double-UCB transplantation, intrabone injection or ex vivo expansion. In this article the authors review the current state of the art, future trends and challenges on the ex vivo expansion of UCB HSCs, focusing on culture parameters affecting the yield and quality of the expanded HSC grafts: novel HSC selection schemes prior to cell culture, cytokine/growth factor cocktails, the impact of biochemical factors (e.g. O2 ) or the addition of supportive cells, e.g. mesenchymal stem/stromal cell (MSC)-based feeder layers) were addressed. Importantly, a critical challenge in cellular therapy is still the scalability, reproducibility and control of the expansion process, in order to meet the clinical requirements for therapeutic applications. Efficient design of bioreactor systems and operation modes are now the focus of many bioengineers, integrating the increasing 'know-how' on HSC biology and physiology, while complying with the GMP standards for the production of cellular products, i.e. through the use of commercially available, highly controlled, disposable technologies.
Collapse
Affiliation(s)
- Pedro Z Andrade
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal.,Cell2b, Advanced Therapeutics, Biocant Park, Cantanhede, Portugal
| | - Francisco dos Santos
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal.,Cell2b, Advanced Therapeutics, Biocant Park, Cantanhede, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal
| |
Collapse
|
32
|
Yu X, Miao J, Xia W, Gu ZJ. Preferential expansion of umbilical cord blood-derived CD34-positive cells on human leukemia inhibitory factor transgenic feeder cells cultured on regenerated silk fibroin film. J Biomed Mater Res B Appl Biomater 2013; 101:964-71. [DOI: 10.1002/jbm.b.32903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 10/29/2012] [Accepted: 01/07/2013] [Indexed: 12/30/2022]
|
33
|
Huang J, Nguyen-McCarty M, Hexner EO, Danet-Desnoyers G, Klein PS. Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nat Med 2012; 18:1778-85. [PMID: 23142822 PMCID: PMC3518679 DOI: 10.1038/nm.2984] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 09/27/2012] [Indexed: 12/14/2022]
Abstract
Hematopoietic stem cell (HSC) self renewal and lineage commitment depend on complex interactions with the microenvironment. The ability to maintain or expand HSCs for clinical applications or basic research has been substantially limited because these interactions are not well defined. Recent evidence suggests that HSCs reside in a low-perfusion, reduced-nutrient niche and that nutrient-sensing pathways contribute to HSC homeostasis. Here we report that suppression of the mTOR pathway, an established nutrient sensor, combined with activation of canonical Wnt-β-catenin signaling, allows for the ex vivo maintenance of human and mouse long-term HSCs under cytokine-free conditions. We also show that the combination of two clinically approved medications that together activate Wnt-β-catenin and inhibit mTOR signaling increases the number (but not the proportion) of long-term HSCs in vivo.
Collapse
Affiliation(s)
- Jian Huang
- Department of Medicine (Hematology-Oncology), University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
34
|
Du J, Wang J, Kong G, Jiang J, Zhang J, Liu Y, Tong W, Zhang J. Signaling profiling at the single-cell level identifies a distinct signaling signature in murine hematopoietic stem cells. Stem Cells 2012; 30:1447-54. [PMID: 22628264 DOI: 10.1002/stem.1127] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cell (HSC) function is tightly regulated by cytokine signaling. Although phospho-flow cytometry allows us to study signaling in defined populations of cells, there has been tremendous hurdle to carry out this study in rare HSCs due to unrecoverable critical HSC markers, low HSC number, and poor cell recovery rate. Here, we overcame these difficulties and developed a "HSC phospho-flow" method to analyze cytokine signaling in murine HSCs at the single-cell level and compare HSC signaling profile to that of multipotent progenitors (MPPs), a cell type immediately downstream of HSCs, and commonly used Lin(-) cKit(+) cells (LK cells, enriched for myeloid progenitors). We chose to study signaling evoked from three representative cytokines, stem cell factor (SCF) and thrombopoietin (TPO) that are essential for HSC function and granulocyte macrophage-colony-stimulating factor (GM-CSF) that is dispensable for HSCs. HSCs display a distinct TPO and GM-CSF signaling signature from MPPs and LK cells, which highly correlates with receptor surface expression. In contrast, although majority of LK cells express lower levels of cKit than HSCs and MPPs, SCF-evoked ERK1/2 activation in LK cells shows a significantly increased magnitude for a prolonged period. These results suggest that specific cellular context plays a more important role than receptor surface expression in SCF signaling. Our study of HSC signaling at the homeostasis stage paves the way to investigate signaling changes in HSCs under conditions of stress, aging, and hematopoietic diseases.
Collapse
Affiliation(s)
- Juan Du
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, WI 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Diogo MM, da Silva CL, Cabral JMS. Separation technologies for stem cell bioprocessing. Biotechnol Bioeng 2012; 109:2699-709. [PMID: 22887094 DOI: 10.1002/bit.24706] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 02/06/2023]
Abstract
Stem cells have been the focus of an intense research due to their potential in Regenerative Medicine, drug discovery, toxicology studies, as well as for fundamental studies on developmental biology and human disease mechanisms. To fully accomplish this potential, the successful application of separation processes for the isolation and purification of stem cells and stem cell-derived cells is a crucial issue. Although separation methods have been used over the past decades for the isolation and enrichment of hematopoietic stem/progenitor cells for transplantation in hemato-oncological settings, recent achievements in the stem cell field have created new challenges including the need for novel scalable separation processes with a higher resolution and more cost-effective. Important examples are the need for high-resolution methods for the separation of heterogeneous populations of multipotent adult stem cells to study their differential biological features and clinical utility, as well as for the depletion of tumorigenic cells after pluripotent stem cell differentiation. Focusing on these challenges, this review presents a critical assessment of separation processes that have been used in the stem cell field, as well as their current and potential applications. The techniques are grouped according to the fundamental principles that govern cell separation, which are defined by the main physical, biophysical, and affinity properties of cells. A special emphasis is given to novel and promising approaches such as affinity-based methods that take advantage of the use of new ligands (e.g., aptamers, lectins), as well as to novel biophysical-based methods requiring no cell labeling and integrated with microscale technologies.
Collapse
Affiliation(s)
- Maria Margarida Diogo
- Department of Bioengineering and Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Technical University of Lisbon, Lisbon, Portugal
| | | | | |
Collapse
|
36
|
RNAi screen identifies MAPK14 as a druggable suppressor of human hematopoietic stem cell expansion. Blood 2012; 119:6255-8. [DOI: 10.1182/blood-2012-01-403949] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We report on a forward RNAi screen in primary human hematopoietic stem and progenitor cells, using pooled lentiviral shRNA libraries deconvoluted by next generation sequencing. We identify MAPK14/p38α as a modulator of ex vivo stem cell proliferation and show that pharmacologic inhibition of p38 dramatically enhances the stem cell activity of cultured umbilical cord blood derived hematopoietic cells. p38 inhibitors should thus be considered in strategies aiming at expanding stem cells for clinical benefit.
Collapse
|
37
|
Holmes T, Yan F, Ko KH, Nordon R, Song E, O'Brien TA, Dolnikov A. Ex vivo expansion of cord blood progenitors impairs their short-term and long-term repopulating activity associated with transcriptional dysregulation of signalling networks. Cell Prolif 2012; 45:266-78. [PMID: 22429797 DOI: 10.1111/j.1365-2184.2012.00813.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 01/13/2012] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Cord blood (CB) has been established to be an alternative source of haematopoietic stem/progenitor cells (HPC) for transplantation. The number of HPC per CB unit is limited, which results in engraftment delay. Ex vivo expansion of HPC improvement must overcome this. MATERIALS AND METHODS Flow cytometry was used to extensively phenotype HPC pre- and post-expansion and CFDA-SE staining was used to track cell divisions. The NSG mouse model was employed in transplantation studies to determine long and short term repopulation in human cells. Gene array analysis was used to evaluate signalling pathways regulated following ex vivo expansion of HPC. RESULTS expansion of CD34(+) HPC impaired their regenerative function. In this xenograft transplantation model we showed that repopulating activity of CB cells declined following expansion. Expanded HPC had delayed engraftment at early and late stages post-transplant. High resolution division tracking revealed that the cultured HPC had reduced expansion and self-renewal probability and increased differentiation rate compared to non-expanded cells. Gene expression analysis exposed significant modulation of a complex network of genes and pathways that normally maintain HPC proliferation and limit their differentiation. CONCLUSIONS The decline in short-term engraftment is consistent with the loss of rapid SCID repopulating ability r(SRA) by expanded CD34(+) CD38(+) cells recently reported. Our data raise concerns for future clinical applications of expanded HPC alone in transplantation.
Collapse
Affiliation(s)
- T Holmes
- Sydney Cord & Marrow Transplant Facility, Sydney Children's Hospital, Sydney, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|
38
|
Drake AC, Chen Q, Chen J. Engineering humanized mice for improved hematopoietic reconstitution. Cell Mol Immunol 2012; 9:215-24. [PMID: 22425741 DOI: 10.1038/cmi.2012.6] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Humanized mice are immunodeficient animals engrafted with human hematopoietic stem cells that give rise to various lineages of human blood cells throughout the life of the mouse. This article reviews recent advances in the generation of humanized mice, focusing on practical considerations. We discuss features of different immunodeficient recipient mouse strains, sources of human hematopoietic stem cells, advances in expansion and genetic modification of hematopoietic stem cells, and techniques to modulate the cytokine environment of recipient mice, in order to enhance reconstitution of specific human blood lineage cells. We highlight the opportunities created by new technologies and discuss practical considerations on how to make best use of the widening array of basic models for specific research applications.
Collapse
Affiliation(s)
- Adam C Drake
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | |
Collapse
|
39
|
Baum C, Modlich U, Göhring G, Schlegelberger B. Concise review: managing genotoxicity in the therapeutic modification of stem cells. Stem Cells 2012; 29:1479-84. [PMID: 21898683 DOI: 10.1002/stem.716] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The therapeutic use of procedures for genetic stem cell modification is limited by potential adverse events related to uncontrolled mutagenesis. Prominent findings have been made in hematopoietic gene therapy, demonstrating the risk of clonal, potentially malignant outgrowth on the basis of mutations acquired during or after therapeutic genome modification. The incidence and the growth rate of insertional mutants have been linked to the "stemness" of the target cells and vector-related features such as the integration pattern, the architecture, and the exact content of transgene cassettes. Milieu factors supporting the survival and expansion of mutants may eventually allow oncogenic progression. Similar concerns apply for medicinal products based on pluripotent stem cells. Focusing on the genetic stress induced by insertional mutagenesis and culture adaptation, we propose four conclusions. (a) Mutations occurring in the production of stem cell-based medicines may be unavoidable and need to be classified according to their risk to trigger the formation of clones that are sufficiently long-lived and mitotically active to acquire secondary transforming mutations. (b) The development of rational prevention strategies depends upon the identification of the specific mutations forming such "dominant clones" (which can also be addressed as cancer stem cell precursors) and a better knowledge of the mechanisms underlying their creation, expansion, and homeostatic control. (c) Quantitative assay systems are required to assess the practical value of preventive actions. (d) Improved approaches for the genetic modification of stem cells can address all critical steps in the origin and growth control of mutants.
Collapse
Affiliation(s)
- Christopher Baum
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
| | | | | | | |
Collapse
|
40
|
Andrade PZ, da Silva CL, dos Santos F, Almeida-Porada G, Cabral JMS. Initial CD34+ cell-enrichment of cord blood determines hematopoietic stem/progenitor cell yield upon ex vivo expansion. J Cell Biochem 2011; 112:1822-31. [PMID: 21400571 DOI: 10.1002/jcb.23099] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Since umbilical cord blood (UCB), contains a limited hematopoietic stem/progenitor cells (HSC) number, successful expansion protocols are needed to overcome the hurdles associated with inadequate numbers of HSC collected for transplantation. UCB cultures were performed using a human stromal-based serum-free culture system to evaluate the effect of different initial CD34(+) cell enrichments (Low: 24 ± 1.8%, Medium: 46 ± 2.6%, and High: 91 ± 1.5%) on the culture dynamics and outcome of HSC expansion. By combining PKH tracking dye with CD34(+) and CD34(+) CD90(+) expression, we have identified early activation of CD34 expression on CD34(-) cells in Low and Medium conditions, prior to cell division (35 ± 4.7% and 55 ± 4.1% CD34(+) cells at day 1, respectively), affecting proliferation/cell cycle status and ultimately determining CD34(+)/CD34(+) CD90(+) cell yield (High: 14 ± 1.0/3.5 ± 1.4-fold; Medium: 22 ± 2.0/3.4 ± 1,0-fold; Low: 31 ± 3.0/4.4 ± 1.5-fold) after a 7-day expansion. Considering the potential benefits of using expanded UCB HSC in transplantation, here we quantified in single UCB units, the impact of using one/two immunomagnetic sorting cycles (corresponding to Medium and High initial progenitor content), and the average CD34(+) cell recovery for each strategy, on overall CD34(+) cell expansion. The higher cell recovery upon one sorting cycle lead to higher CD34(+) cell numbers after 7 days of expansion (30 ± 2.0 vs. 13 ± 1.0 × 10(6) cells). In particular, a high (>90%) initial progenitor content was not mandatory to successfully expand HSC, since cell populations with moderate levels of enrichment readily increased CD34 expression ex-vivo, generating higher stem/progenitor cell yields. Overall, our findings stress the importance of establishing a balance between the cell proliferative potential and cell recovery upon purification, towards the efficient and cost-effective expansion of HSC for cellular therapy.
Collapse
Affiliation(s)
- Pedro Z Andrade
- Department of Bioengineering, Instituto Superior Técnico, Lisboa, Portugal
| | | | | | | | | |
Collapse
|
41
|
Reduced-intensity conditioning with combined haploidentical and cord blood transplantation results in rapid engraftment, low GVHD, and durable remissions. Blood 2011; 118:6438-45. [PMID: 21976674 DOI: 10.1182/blood-2011-08-372508] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We conducted a 45 patient prospective study of reduced-intensity conditioning (RIC) and transplantation of unrelated umbilical cord blood (UCB) and CD34(+) stem cells from a haploidentical family member. Median age was 50 years; weight was 80 kg. Fifty-eight percent had active disease. Neutrophil engraftment occurred at 11 days (interquartile range [IQR], 9-15) and platelet engraftment at 19 days (IQR, 15-33). In the majority of patients, early haploidentical engraftment was replaced by durable engraftment of UCB by 100 days, with regular persistence of minor host and/or haplo-hematopoiesis. Percentage of haplochimerism at day 100 correlated with the haplo-CD34 dose (P = .003). Cumulative incidence of acute GVHD (aGVHD) was 25% and chronic GVHD (cGVHD) was 5%. Actuarial survival at 1 year was 55%, progression-free survival (PFS) was 42%, nonrelapse mortality (NRM) was 28%, and relapse was 30%. RIC and haplo-cord transplantation results in fast engraftment of neutrophils and platelets, low incidences of aGVHD and cGVHD, low frequency of delayed opportunistic infections, reduced transfusion requirements, shortened length of hospital stay, and promising long-term outcomes. UCB cell dose had no impact on time to hematopoietic recovery. Therefore, UCB selection can prioritize matching, and better matched donors can be identified rapidly for most patients. This study is registered at http://clinicaltrials.gov as NCI clinical trial no. NCT00943800.
Collapse
|
42
|
Perry JM, He XC, Sugimura R, Grindley JC, Haug JS, Ding S, Li L. Cooperation between both Wnt/{beta}-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev 2011; 25:1928-42. [PMID: 21890648 DOI: 10.1101/gad.17421911] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although self-renewal is the central property of stem cells, the underlying mechanism remains inadequately defined. Using a hematopoietic stem and progenitor cell (HSPC)-specific conditional induction line, we generated a compound genetic model bearing both Pten deletion and β-catenin activation. These double mutant mice exhibit a novel phenotype, including expansion of phenotypic long-term hematopoietic stem cells (LT-HSCs) without extensive differentiation. Unexpectedly, constitutive activation of β-catenin alone results in apoptosis of HSCs. However, together, the Wnt/β-catenin and PTEN/PI3k/Akt pathways interact to drive phenotypic LT-HSC expansion by inducing proliferation while simultaneously inhibiting apoptosis and blocking differentiation, demonstrating the necessity of complementary cooperation between the two pathways in promoting self-renewal. Mechanistically, β-catenin activation reduces multiple differentiation-inducing transcription factors, blocking differentiation partially through up-regulation of Inhibitor of differentiation 2 (Id2). In double mutants, loss of Pten enhances the HSC anti-apoptotic factor Mcl-1. All of these contribute in a complementary way to HSC self-renewal and expansion. While permanent, genetic alteration of both pathways in double mutant mice leads to expansion of phenotypic HSCs, these HSCs cannot function due to blocked differentiation. We developed a pharmacological approach to expand normal, functional HSCs in culture using factors that reversibly activate both Wnt/β-catenin and PI3K/Akt signaling simultaneously. We show for the first time that activation of either single pathway is insufficient to expand primitive HSCs, but in combination, both pathways drive self-renewal and expansion of HSCs with long-term functional capacity.
Collapse
Affiliation(s)
- John M Perry
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Sousa AF, Andrade PZ, Pirzgalska RM, Galhoz TM, Azevedo AM, da Silva CL, Aires-Barros MR, Cabral JMS. A novel method for human hematopoietic stem/progenitor cell isolation from umbilical cord blood based on immunoaffinity aqueous two-phase partitioning. Biotechnol Lett 2011; 33:2373-7. [PMID: 21858668 DOI: 10.1007/s10529-011-0727-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 08/05/2011] [Indexed: 11/27/2022]
Abstract
A novel cell separation process based on immunoaffinity aqueous two phase systems is presented to isolate and purify CD34(+) stem/progenitor cells directly from the whole umbilical cord blood (UCB). A system, composed of polyethylene glycol and dextran, was evaluated for the selective recovery of CD34(+) cells from UCB. A monoclonal antibody against the CD34 surface antigen was used for the direct partitioning of CD34(+) cells in UCB to the PEG-rich phase. The initial population of CD34(+) cells (0.2% of the initial sample) was enriched to values up to 42% in a single partitioning step, while the majority of contaminant cells were partitioned to the dextran-rich phase (1.37 × 10(-2) < K(P) < 2.76 × 10(-2)). This novel selection method allowed a recovery yield of 95% of CD34(+) cells with a purification factor of 245 and is expected to pave a new way to purify hematopoietic stem/progenitor cells for use in a variety of clinical settings.
Collapse
Affiliation(s)
- António F Sousa
- Department of Bioengineering, Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Technical University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Koen van Besien
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
45
|
Abstract
Despite progress in our understanding of the growth factors that support the progressive maturation of the various cell lineages of the hematopoietic system, less is known about factors that govern the self-renewal of hematopoietic stem and progenitor cells (HSPCs), and our ability to expand human HSPC numbers ex vivo remains limited. Interest in stem cell expansion has been heightened by the increasing importance of HSCs in the treatment of both malignant and nonmalignant diseases, as well as their use in gene therapy. To date, most attempts to ex vivo expand HSPCs have used hematopoietic growth factors but have not achieved clinically relevant effects. More recent approaches, including our studies in which activation of the Notch signaling pathway has enabled a clinically relevant ex vivo expansion of HSPCs, have led to renewed interest in this arena. Here we briefly review early attempts at ex vivo expansion by cytokine stimulation followed by an examination of our studies investigating the role of Notch signaling in HSPC self-renewal. We will also review other recently developed approaches for ex vivo expansion, primarily focused on the more extensively studied cord blood-derived stem cell. Finally, we discuss some of the challenges still facing this field.
Collapse
|