1
|
Ding L, Oh S, Shrestha J, Lam A, Wang Y, Radfar P, Warkiani ME. Scaling up stem cell production: harnessing the potential of microfluidic devices. Biotechnol Adv 2023; 69:108271. [PMID: 37844769 DOI: 10.1016/j.biotechadv.2023.108271] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Stem cells are specialised cells characterised by their unique ability to both self-renew and transform into a wide array of specialised cell types. The widespread interest in stem cells for regenerative medicine and cultivated meat has led to a significant demand for these cells in both research and practical applications. Despite the growing need for stem cell manufacturing, the industry faces significant obstacles, including high costs for equipment and maintenance, complicated operation, and low product quality and yield. Microfluidic technology presents a promising solution to the abovementioned challenges. As an innovative approach for manipulating liquids and cells within microchannels, microfluidics offers a plethora of advantages at an industrial scale. These benefits encompass low setup costs, ease of operation and multiplexing, minimal energy consumption, and the added advantage of being labour-free. This review presents a thorough examination of the prominent microfluidic technologies employed in stem cell research and explores their promising applications in the burgeoning stem cell industry. It thoroughly examines how microfluidics can enhance cell harvesting from tissue samples, facilitate mixing and cryopreservation, streamline microcarrier production, and efficiently conduct cell separation, purification, washing, and final cell formulation post-culture.
Collapse
Affiliation(s)
- Lin Ding
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia.
| | - Steve Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Alan Lam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Yaqing Wang
- School of Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Payar Radfar
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia..
| |
Collapse
|
2
|
Vašíček J, Shehata M, Schnabl S, Hilgarth M, Hubmann R, Jäger U, Bauer M, Chrenek P. Critical assessment of the efficiency of CD34 and CD133 antibodies for enrichment of rabbit hematopoietic stem cells. Biotechnol Prog 2018; 34:1278-1289. [PMID: 29882300 DOI: 10.1002/btpr.2659] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 03/25/2018] [Indexed: 12/11/2022]
Abstract
Rabbits have many hereditary diseases common to humans and are therefore a valuable model for regenerative disease and hematopoietic stem cell (HSC) therapies. Currently, there is no substantial data on the isolation and/or enrichment of rabbit HSCs. This study was initiated to evaluate the efficiency of the commercially available anti-CD34 and anti-CD133 antibodies for the detection and potential enrichment of rabbit HSCs from peripheral blood. PBMCs from rabbit and human blood were labelled with different clones of anti-human CD34 monoclonal antibodies (AC136, 581, and 8G12) and rabbit polyclonal CD34 antibody (pCD34) and anti-human CD133 monoclonal antibodies (AC133 and 293C3). Flow cytometry showed a higher percentage of rabbit CD34+ cells labelled by AC136 in comparison to the clone 581 and pCD34 (P < 0.01). A higher percentage of rabbit CD133+ cells were also detected by 293C3 compared to the AC133 clone (P < 0.01). Therefore, AC136 clone was used for the indirect immunomagnetic enrichment of rabbit CD34+ cells using magnetic-activated cell sorting (MACS). The enrichment of the rabbit CD34+ cells after sorting was low in comparison to human samples (2.4% vs. 39.6%). PCR analyses confirmed the efficient enrichment of human CD34+ cells and the low expression of CD34 mRNA in rabbit positive fraction. In conclusion, the tested antibodies might be suitable for detection, but not for sorting the rabbit CD34+ HSCs and new specific anti-rabbit CD34 antibodies are needed for efficient enrichment of rabbit HSCs. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 2018 © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1278-1289, 2018.
Collapse
Affiliation(s)
- Jaromír Vašíček
- NAFC-Research Institute for Animal Production in Nitra, Institute of Farm Animal Genetics and Reproduction, Lužianky, Slovak Republic, Hlohovecká 2, 951 41.,Research Centre AgroBioTech, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic.,Faculty of Biotechnology and Food Science, Department of Biochemistry and Biotechnology, Slovak University of Agriculture, Nitra, Tr A. Hlinku 2, 949 76, Slovak Republic
| | - Medhat Shehata
- Dept. of Internal Medicine I, Div. of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Susanne Schnabl
- Dept. of Internal Medicine I, Div. of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Martin Hilgarth
- Dept. of Internal Medicine I, Div. of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Rainer Hubmann
- Dept. of Internal Medicine I, Div. of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Ulrich Jäger
- Dept. of Internal Medicine I, Div. of Haematology and Haemostaseology, Comprehensive Cancer Centre Vienna, Drug and Target Screening Unit DTSU, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Miroslav Bauer
- NAFC-Research Institute for Animal Production in Nitra, Institute of Farm Animal Genetics and Reproduction, Lužianky, Slovak Republic, Hlohovecká 2, 951 41.,Faculty of Natural Sciences, Department of Botany and Genetics, Constantine the Philosopher University in Nitra, 949 74 Nitra, mládeže, Slovak Republic, Nábrežie 91
| | - Peter Chrenek
- NAFC-Research Institute for Animal Production in Nitra, Institute of Farm Animal Genetics and Reproduction, Lužianky, Slovak Republic, Hlohovecká 2, 951 41.,Faculty of Biotechnology and Food Science, Department of Biochemistry and Biotechnology, Slovak University of Agriculture, Nitra, Tr A. Hlinku 2, 949 76, Slovak Republic
| |
Collapse
|
3
|
Painter MM, Zaikos TD, Collins KL. Quiescence Promotes Latent HIV Infection and Resistance to Reactivation from Latency with Histone Deacetylase Inhibitors. J Virol 2017; 91:e01080-17. [PMID: 29021396 PMCID: PMC5709582 DOI: 10.1128/jvi.01080-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/02/2017] [Indexed: 12/28/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) establishes transcriptionally silent latent infections in resting memory T cells and hematopoietic stem and progenitor cells (HSPCs), which allows the virus to persist in infected individuals despite antiretroviral therapy. Developing in vitro models of HIV-1 latency that recapitulate the characteristics of latently infected cells in vivo is crucial to identifying and developing effective latency-reversing therapies. HSPCs exist in a quiescent state in vivo, and quiescence is correlated with latent infections in T cells. However, current models for culturing HSPCs and for infecting T cells in vitro require that the cells be maintained in an actively proliferating state. Here we describe a novel culture system in which primary human HSPCs cultured under hypothermic conditions are maintained in a quiescent state. We show that these quiescent HSPCs are susceptible to predominantly latent infection with HIV-1, while actively proliferating and differentiating HSPCs obtain predominantly active infections. Furthermore, we demonstrate that the most primitive quiescent HSPCs are more resistant to spontaneous reactivation from latency than more differentiated HSPCs and that quiescent HSPCs are resistant to reactivation by histone deacetylase inhibitors or P-TEFb activation but are susceptible to reactivation by protein kinase C (PKC) agonists. We also demonstrate that inhibition of HSP90, a known regulator of HIV transcription, recapitulates the quiescence and latency phenotypes of hypothermia, suggesting that hypothermia and HSP90 inhibition may regulate these processes by similar mechanisms. In summary, these studies describe a novel model for studying HIV-1 latency in human primary cells maintained in a quiescent state.IMPORTANCE Human immunodeficiency virus type 1 (HIV-1) establishes a persistent infection for which there remains no feasible cure. Current approaches are unable to clear the virus despite decades of therapy due to the existence of latent reservoirs of integrated HIV-1, which can reactivate and contribute to viral rebound following treatment interruption. Previous clinical attempts to reactivate the latent reservoirs in an individual so that they can be eliminated by the immune response or viral cytopathic effect have failed, indicating the need for a better understanding of the processes regulating HIV-1 latency. Here we characterize a novel in vitro model of HIV-1 latency in primary hematopoietic stem and progenitor cells isolated from human cord blood that may better recapitulate the behavior of latently infected cells in vivo This model can be used to study mechanisms regulating latency and potential therapeutic approaches to reactivate latent infections in quiescent cells.
Collapse
Affiliation(s)
- Mark M Painter
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas D Zaikos
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathleen L Collins
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Zhang Q, Yin T, Xu R, Gao W, Zhao H, Shapter JG, Wang K, Shen Y, Huang P, Gao G, Wu Y, Cui D. Large-scale immuno-magnetic cell sorting of T cells based on a self-designed high-throughput system for potential clinical application. NANOSCALE 2017; 9:13592-13599. [PMID: 28875998 DOI: 10.1039/c7nr04914e] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this contribution, we designed four types of immuno-magnetic nanoparticles for separation of different T cells (CD3+, CD4+, CD8+ and CD14+ T cells), and we established a new large-scale immuno-magnetic cell sorting procedure to achieve an enrichment of particular T cells using our designed auto-IMACS device. This device could achieve recyclable large-scale cell sorting, for which the throughput of the system reached ∼4000 mL and the maximum cell capacity was 4 × 1010. The collected cells were analyzed by flow cytometry and visual cytology data, and the effective selection rates of CD3+, CD4+, CD8+ and CD14+ T cells were 79.3%, 74.1%, 57.1% and 67.9%, respectively. The sorted CD8+ T cells still retained good cytotoxic activity against specific cells. In addition, the sorted T cells can also be further incubated in vitro and proliferated, and even could be infused back into patients for immunotherapy in the near future.
Collapse
Affiliation(s)
- Qian Zhang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent diagnosis and treatment instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China.
| | - Ting Yin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent diagnosis and treatment instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China.
| | - Rongrong Xu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai 200433, China.
| | - Wenjun Gao
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
| | - Hui Zhao
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
| | - Joseph G Shapter
- School of Chemical and Physical Sciences, Flinders University, Bedford Park, Adelaide 5042, Australia
| | - Kan Wang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent diagnosis and treatment instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China.
| | - Yulan Shen
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai JiaoTong University, Shanghai, 200240, China
| | - Peng Huang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent diagnosis and treatment instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China.
| | - Guo Gao
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent diagnosis and treatment instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China.
| | - Yanfeng Wu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai 200433, China.
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent diagnosis and treatment instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China.
| |
Collapse
|
5
|
Tunneling nanotubes mediate the transfer of stem cell marker CD133 between hematopoietic progenitor cells. Exp Hematol 2016; 44:1092-1112.e2. [PMID: 27473566 DOI: 10.1016/j.exphem.2016.07.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/15/2016] [Accepted: 07/16/2016] [Indexed: 12/24/2022]
Abstract
Deciphering all mechanisms of intercellular communication used by hematopoietic progenitors is important, not only for basic stem cell research, but also in view of their therapeutic relevance. Here, we investigated whether these cells can produce the thin F-actin-based plasma membrane protrusions referred to as tunneling nanotubes (TNTs), which are known to bridge cells over long distances without contact with the substratum and transfer cargo molecules along them in various biological processes. We found that human primary CD34+ hematopoietic progenitors and leukemic KG1a cells develop such structures upon culture on primary mesenchymal stromal cells or specific extracellular-matrix-based substrata. Time-lapse video microscopy revealed that cell dislodgement is the primary mechanism responsible for TNT biogenesis. Surprisingly, we found that, among various cluster of differentiation (CD) markers, only the stem cell antigen CD133 is transferred between cells. It is selectively and directionally transported along the surface of TNTs in small clusters, such as cytoplasmic phospho-myosin light chain 2, suggesting that the latter actin motor protein might be implicated in this process. Our data provide new insights into the biology of hematopoietic progenitors that can contribute to our understanding of all facets of intercellular communication in the bone marrow microenvironment under healthy or cancerous conditions.
Collapse
|
6
|
Reichert D, Friedrichs J, Ritter S, Käubler T, Werner C, Bornhäuser M, Corbeil D. Phenotypic, Morphological and Adhesive Differences of Human Hematopoietic Progenitor Cells Cultured on Murine versus Human Mesenchymal Stromal Cells. Sci Rep 2015; 5:15680. [PMID: 26498381 PMCID: PMC4620509 DOI: 10.1038/srep15680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023] Open
Abstract
Xenogenic transplantation models have been developed to study human hematopoiesis in immunocompromised murine recipients. They still have limitations and therefore it is important to delineate all players within the bone marrow that could account for species-specific differences. Here, we evaluated the proliferative capacity, morphological and physical characteristics of human CD34+ hematopoietic stem and progenitor cells (HSPCs) after co-culture on murine or human bone marrow-derived mesenchymal stromal cells (MSCs). After seven days, human CD34+CD133– HSPCs expanded to similar extents on both feeder layers while cellular subsets comprising primitive CD34+CD133+ and CD133+CD34– phenotypes are reduced fivefold on murine MSCs. The number of migrating HSPCs was also reduced on murine cells suggesting that MSC adhesion influences cellular polarization of HSPC. We used atomic force microscopy-based single-cell force spectroscopy to quantify their adhesive interactions. We found threefold higher detachment forces of human HSPCs from murine MSCs compared to human ones. This difference is related to the N-cadherin expression level on murine MSCs since its knockdown abolished their differential adhesion properties with human HSPCs. Our observations highlight phenotypic, morphological and adhesive differences of human HSPCs when cultured on murine or human MSCs, which raise some caution in data interpretation when xenogenic transplantation models are used.
Collapse
Affiliation(s)
- Doreen Reichert
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Jens Friedrichs
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Steffi Ritter
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Theresa Käubler
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany
| | - Carsten Werner
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, 01307 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| | - Denis Corbeil
- Tissue Engineering Laboratories (BIOTEC), Technische Universität Dresden, 01307 Dresden, Germany.,DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden 01307 Dresden, Germany
| |
Collapse
|
7
|
Shen Y, Ren X, Ding K, Zhang Z, Wang D, Pan J. Antitumor activity of S116836, a novel tyrosine kinase inhibitor, against imatinib-resistant FIP1L1-PDGFRα-expressing cells. Oncotarget 2015; 5:10407-20. [PMID: 25431951 PMCID: PMC4279382 DOI: 10.18632/oncotarget.2090] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/10/2014] [Indexed: 01/22/2023] Open
Abstract
The FIP1-like-1-platelet-derived growth factor receptor alpha (FIP1L1-PDGFRα) fusion oncogene is the driver factor in a subset of patients with hypereosinophilic syndrome (HES)/chronic eosinophilic leukemia (CEL). Most FIP1L1-PDGFRα-positive patients respond well to the tyrosine kinase inhibitor (TKI) imatinib. Resistance to imatinib in HES/CEL has been described mainly due to the T674I mutation in FIP1L1-PDGFRα, which is homologous to the imatinib-resistant T315I mutation in BCR-ABL. Development of novel TKIs is imperative to overcome resistance to imatinib. We synthesized S116836, a novel TKI. In this study, we evaluated the antitumor activity of S116836 in FIP1L1-PDGFRα-expressing cells. The results showed that S116836 potently inhibited PDGFRα and its downstream signaling molecules such as STAT3, AKT, and Erk1/2. S116836 effectively inhibited the growth of the WT and T674I FIP1L1-PDGFRα-expressing neoplastic cells in vitro and in nude mouse xenografts. Moreover, S116836 induced intrinsic pathway of apoptosis as well as the death receptor pathway, coincided with up-regulation of the proapoptotic BH3-only protein Bim-EL through the Erk1/2 pathway. In conclusion, S116836 is active against WT and T674I FIP1L1-PDGFRα-expressing cells, and may be a prospective agent for the treatment of HES/CEL.
Collapse
Affiliation(s)
- Yingying Shen
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaomei Ren
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ke Ding
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhang Zhang
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Deping Wang
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jingxuan Pan
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China. Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
8
|
Ohlfest JR, Zellmer DM, Panyam J, Swaminathan SK, Oh S, Waldron NN, Toma S, Vallera DA. Immunotoxin targeting CD133(+) breast carcinoma cells. Drug Deliv Transl Res 2015; 3:195-204. [PMID: 25787984 DOI: 10.1007/s13346-012-0066-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
CD133 expression enriches for tumor-initiating cells and is a negative prognostic factor in numerous cancers. We previously developed an immunotoxin against CD133 by fusing a gene fragment encoding the scFv portion of an anti-CD133 antibody to a gene fragment encoding deimmunized PE38KDEL. The resulting fusion protein, dCD133KDEL, demonstrated potent antitumor activity following intratumoral delivery into head neck cell carcinoma xenografts. However, the efficacy against other tumors and the tolerability of systemic administration remained unclear. The purpose of this study was to evaluate the tolerability and efficacy of dCD133KDEL in a systemic human breast carcinoma model. Time course viability studies showed that dCD133KDEL selectively inhibited MDA-MB-231 ductal breast carcinoma cells that contained a minority CD133(+) subpopulation, implicating CD133(+) cells as a source for self-renewal within this cell line. Furthermore, systemic administration of dCD133KDEL caused regression or inhibition of tumor growth in mice bearing an intrasplenic MDA-MB-231 tumor challenge as a model for metastatic disease. In the same model, combined therapy with dCD133KDEL and another immunotoxin designed to target the bulk tumor mass was the most effective therapy, supporting the idea that such combination therapies might better address tumor heterogeneity. dCD133KDEL shows promise as a therapeutic agent and as a biologic tool to study cancer stem cells.
Collapse
Affiliation(s)
- John R Ohlfest
- Department of Pediatrics, Masonic Cancer Center of the University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Brendel C, Goebel B, Daniela A, Brugman M, Kneissl S, Schwäble J, Kaufmann KB, Müller-Kuller U, Kunkel H, Chen-Wichmann L, Abel T, Serve H, Bystrykh L, Buchholz CJ, Grez M. CD133-targeted gene transfer into long-term repopulating hematopoietic stem cells. Mol Ther 2014; 23:63-70. [PMID: 25189742 DOI: 10.1038/mt.2014.173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/25/2014] [Indexed: 11/09/2022] Open
Abstract
Gene therapy for hematological disorders relies on the genetic modification of CD34(+) cells, a heterogeneous cell population containing about 0.01% long-term repopulating cells. Here, we show that the lentiviral vector CD133-LV, which uses a surface marker on human primitive hematopoietic stem cells (HSCs) as entry receptor, transfers genes preferentially into cells with high engraftment capability. Transduction of unstimulated CD34(+) cells with CD133-LV resulted in gene marking of cells with competitive proliferative advantage in vitro and in immunodeficient mice. The CD133-LV-transduced population contained significantly more cells with repopulating capacity than cells transduced with vesicular stomatitis virus (VSV)-LV, a lentiviral vector pseudotyped with the vesicular stomatitis virus G protein. Upon transfer of a barcode library, CD133-LV-transduced cells sustained gene marking in vivo for a prolonged period of time with a 6.7-fold higher recovery of barcodes compared to transduced control cells. Moreover, CD133-LV-transduced cells were capable of repopulating secondary recipients. Lastly, we show that this targeting strategy can be used for transfer of a therapeutic gene into CD34(+) cells obtained from patients suffering of X-linked chronic granulomatous disease. In conclusion, direct gene transfer into CD133(+) cells allows for sustained long-term engraftment of gene corrected cells.
Collapse
Affiliation(s)
- Christian Brendel
- 1] Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany [2] Current address: Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin Goebel
- Department of Medicine, Hematology/Oncology, JW-Goethe-University, Frankfurt/M, Germany
| | - Abriss Daniela
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Martijn Brugman
- Department of Immunohematology and Blood Transfusion (IHB) Leiden University Medical Center, Leiden, Netherlands
| | - Sabrina Kneissl
- 1] Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany [2] Current address: Division of Haematology/Transfusion Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - Joachim Schwäble
- Institute for Transfusion Medicine and Immune Hematology, Clinics of the Johann Wolfgang Goethe University, German Red Cross Blood Donor Service Baden-Wuerttemberg-Hessen, Frankfurt am Main, Hessen, Germany
| | - Kerstin B Kaufmann
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Uta Müller-Kuller
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Hana Kunkel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Linping Chen-Wichmann
- 1] Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany [2] Current address: Experimental Cell Therapy and Hematology, Department of Transfusion Medicine, Cell Therapy and Haemostaseology, Ludwig Maximilian University Hospital Munich, Munich, Germany
| | - Tobias Abel
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Hubert Serve
- Department of Medicine, Hematology/Oncology, JW-Goethe-University, Frankfurt/M, Germany
| | - Leonid Bystrykh
- Department of Cell Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Manuel Grez
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| |
Collapse
|
10
|
Indumathi S, Harikrishnan R, Rajkumar JS, Dhanasekaran M. Immunophenotypic comparison of heterogenous non-sorted versus sorted mononuclear cells from human umbilical cord blood: a novel cell enrichment approach. Cytotechnology 2013; 67:107-14. [PMID: 24357150 DOI: 10.1007/s10616-013-9663-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/21/2013] [Indexed: 12/11/2022] Open
Abstract
Human umbilical cord blood (hUCB) has been the preferred source of stem cells for the treatment of haematological malignancies and genetic disorders. This is primarily due to its non-invasiveness, high accessibility with relative ease of isolation. Still failures do prevail due to its heterogeneity and lesser frequency of MSC identified in UCB. This study, thus, employs a cell enrichment technology to improve its therapeutic efficacy. This was achieved by immunophenotypic comparison of stem cells isolated from the heterogenous non-sorted mononuclear cells (MNCs), linage depleted (Lin+ and Lin-) fractions obtained from magnetic activated cell sorter (MACS) and sorted MNCs obtained by fluorescent activated cell sorter (FACS). The markers under consideration were CD29, CD44, CD34, CD45, CD133, CD90 and CD117. FACS sorted MNCs were rich in naive stem cell population, whereas non-sorted MNCs and lineage depleted fractions were found to be rich in progenitors. Thus, we suggest that a combination therapy of both sorted population might serve as an alternative valuable tool in treating haematologic/genetic disorders. However, further research on cell enrichment technology might give a clue for improved cell based therapy in regenerative medicine.
Collapse
Affiliation(s)
- S Indumathi
- Department of Stem Cells, Lifeline RIGID Hospital, Chennai, 600 096, India
| | | | | | | |
Collapse
|
11
|
CD133 is a modifier of hematopoietic progenitor frequencies but is dispensable for the maintenance of mouse hematopoietic stem cells. Proc Natl Acad Sci U S A 2013; 110:5582-7. [PMID: 23509298 DOI: 10.1073/pnas.1215438110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Pentatransmembrane glycoprotein prominin-1 (CD133) is expressed at the cell surface of multiple somatic stem cells, and it is widely used as a cell surface marker for the isolation and characterization of human hematopoietic stem cells (HSCs) and cancer stem cells. CD133 has been linked on a cell biological basis to stem cell-fate decisions in human HSCs and emerges as an important physiological regulator of stem cell maintenance and expansion. Its expression and physiological relevance in the murine hematopoietic system is nevertheless elusive. We show here that CD133 is expressed by bone marrow-resident murine HSCs and myeloid precursor cells with the developmental propensity to give rise to granulocytes and monocytes. However, CD133 is dispensable for the pool size and function of HSCs during steady-state hematopoiesis and after transplantation, demonstrating a substantial species difference between mouse and man. Blood cell numbers in the periphery are normal; however, CD133 appears to be a modifier for the development of growth-factor responsive myeloerythroid precursor cells in the bone marrow under steady state and mature red blood cells after hematopoietic stress. Taken together, these studies show that CD133 is not a critical regulator of hematopoietic stem cell function in mouse but that it modifies frequencies of growth-factor responsive hematopoietic progenitor cells during steady state and after myelotoxic stress in vivo.
Collapse
|
12
|
|
13
|
Handgretinger R, Kuçi S. CD133-Positive Hematopoietic Stem Cells: From Biology to Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 777:99-111. [PMID: 23161078 DOI: 10.1007/978-1-4614-5894-4_7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lifelong hematopoiesis is sustained by a very small number of hematopoietic stem cells capable of self-renewal and differentiation into multiple hematopoietic lineages. The sialomucin CD34 has been, and is currently, used for the identification and purification of primitive hematopoietic progenitors. Depending on the source of stem cells, CD34 may not be expressed on all progenitor cells. An alternative stem cell marker is prominin-1 (CD133), which is expressed on a subpopulation of CD34(+) cells as well as on CD34(-) progenitor cells derived from various sources including fetal liver and bone marrow, adult bone marrow, cord blood, and mobilized peripheral blood. CD133(+) stem cells can reconstitute myelo- and lymphopoiesis of lethally irradiated mice, and the characterization of the CD133 expression on stem cells provides some insights into the biology of the hierarchy and functional organization of human hematopoiesis. The availability of methods for clinical large-scale isolation of CD133(+) cells facilitates their use in autologous and allogeneic hematopoietic stem cell transplantation and possibly in other fields of regenerative medicine.
Collapse
Affiliation(s)
- Rupert Handgretinger
- University Children's Hospital, Department of Hematology/Oncology, Hoppe-Seyler-Strasse 1, 72076, Tübingen, Germany,
| | | |
Collapse
|
14
|
Fonseca AV, Corbeil D. The hematopoietic stem cell polarization and migration: A dynamic link between RhoA signaling pathway, microtubule network and ganglioside-based membrane microdomains. Commun Integr Biol 2011; 4:201-4. [PMID: 21655440 DOI: 10.4161/cib.4.2.14419] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 12/06/2010] [Indexed: 11/19/2022] Open
Abstract
The polarization and migration of eukaryotic cells are fundamental processes for the development and maintenance of a tissue. These aspects gain especial interest when it comes to stem and progenitor cells in the way that their manipulation might open new avenues in regenerative therapy. In recent years, novel biological facets of migrating hematopoietic stem cells were revealed by several groups, including ours. Among these features, the polarization of their membranous (proteins and lipids) and cytoplasmic constituents, which leads to the formation of a specialized sub-cellular structure located at the rear pole-the uropod-has gained increasing interest. In a new study we have demonstrated that such phenomena involve a coordinated mechanism between Rho GTPase signaling and the microtubule network. Specifically, our results based on the use of synthetic inhibitors and RNA interference suggest that the activity of RhoA and its effector ROCK I is indispensable for cell polarization and the active reorganization of microtubules that are required for migration.
Collapse
Affiliation(s)
- Ana-Violeta Fonseca
- Tissue Engineering Laboratories (BIOTEC) and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD); Technische Universität Dresden; Dresden, Germany
| | | |
Collapse
|
15
|
Dendritic cells with lymphocyte-stimulating activity differentiate from human CD133 positive precursors. Blood 2011; 117:3983-95. [PMID: 21304102 DOI: 10.1182/blood-2010-08-299735] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
CD133 is a hallmark of primitive myeloid progenitors. We have addressed whether human cord blood cells selected for CD133 can generate dendritic cells, and Langerhans cells in particular, in conditions that promote that generation from CD34(+) progenitors. Transforming growth factor-β1 (TGF-β1) and anti-TGF-β1 antibody, respectively, were added in some experiments. With TGF-β, monocytoid cells were recognized after 7 days. Immunophenotypically immature dendritic cells were present at day 14. After 4 more days, the cells expressed CD54, CD80, CD83, and CD86 and were potent stimulators in mixed lymphocyte reaction; part of the cells expressed CD1a and langerin, but not Birbeck granules. Without TGF-β, only a small fraction of cells acquired a dendritic shape and expressed the maturation-related antigens, and lymphocytes were poorly stimulated. With anti-TGF-β, the cell growth was greatly hampered, CD54 and langerin were never expressed, and lymphocytes were stimulated weakly. In conclusion, CD133(+) progenitors can give rise in vitro, through definite steps, to mature, immunostimulatory dendritic cells with molecular features of Langerhans cells, although without Birbeck granules. Addition of TGF-β1 helps to stimulate cell growth and promotes the acquisition of mature immunophenotypical and functional features. Neither langerin nor Birbeck granules proved indispensable for lymphocyte stimulation.
Collapse
|
16
|
Sobkow L, Seib FP, Prodanov L, Kurth I, Drichel J, Bornhäuser M, Werner C. Prolonged transendothelial migration of human haematopoietic stem and progenitor cells (HSPCs) towards hydrogel-released SDF1. Ann Hematol 2011; 90:865-71. [DOI: 10.1007/s00277-011-1155-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 01/03/2011] [Indexed: 12/19/2022]
|
17
|
CD49d blockade by natalizumab in patients with multiple sclerosis affects steady-state hematopoiesis and mobilizes progenitors with a distinct phenotype and function. Bone Marrow Transplant 2010; 45:1489-96. [DOI: 10.1038/bmt.2009.381] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Koutna I, Peterkova M, Simara P, Stejskal S, Tesarova L, Kozubek M. Proliferation and differentiation potential of CD133+ and CD34+ populations from the bone marrow and mobilized peripheral blood. Ann Hematol 2010; 90:127-37. [PMID: 20821012 DOI: 10.1007/s00277-010-1058-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 08/17/2010] [Indexed: 01/14/2023]
Abstract
CD34 is the most frequently used marker for the selection of cells for bone marrow (BM) transplantation. The use of CD133 as an alternative marker is an open research topic. The goal of this study was to evaluate the proliferation and differentiation potential for hematopoiesis (short and long term) of CD133+ and CD34+ populations from bone marrow and mobilized peripheral blood. Eight cell populations were compared: CD34+ and CD133+ cells from both the BM (CML Ph-, CML Ph+, and healthy volunteers) and mobilized peripheral blood cells. Multicolor flow cytometry and cultivation experiments were used to measure expression and differentiation of the individual populations. It was observed that the CD133+ BM population showed higher cell expansion. Another finding is that during a 6-day cultivation with 5(6)-carboxyfluorescein diacetate N-succinimidyl ester (CFSE), more cells remained in division D0 (non-dividing cells). There was a higher percentage of CD38- cells observed on the CD133+ BM population. It was also observed that the studied populations contained very similar but not the same pools of progenitors: erythroid, lymphoid, and myeloid. This was confirmed by CFU-GM and CFU-E experiments. The VEGFR antigen was used to monitor subpopulations of endothelial sinusoidal progenitors. The CD133+ BM population contained significantly more VEGFR+ cells. Our findings suggest that the CD133+ population from the BM shows better proliferation activity and a higher distribution of primitive progenitors than any other studied population.
Collapse
Affiliation(s)
- Irena Koutna
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Botanicka 68a, 602 00, Brno, Czech Republic.
| | | | | | | | | | | |
Collapse
|
19
|
Freund D, Fonseca AV, Janich P, Bornhäuser M, Corbeil D. Differential expression of biofunctional GM1 and GM3 gangliosides within the plastic-adherent multipotent mesenchymal stromal cell population. Cytotherapy 2010; 12:131-42. [PMID: 20196693 DOI: 10.3109/14653240903476438] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS It is unclear whether the plastic-adherent multipotent mesenchymal stromal cells (MSC) isolated from human bone marrow (BM) represent a uniform cell population or are heterogeneous in terms of cell-surface constituents and hence functionality. METHODS We investigated the expression profile of certain biofunctional lipids by plastic-adherent MSC, focusing particularly on two membrane microdomain (lipid raft)-associated monosialogangliosides, GM1 and GM3, using indirect confocal laser scanning fluorescence microscopy and flow cytometry. RESULTS Phenotypically, we observed a differential expression where certain MSC subsets exhibited GM1, GM3 or both at the plasma membrane. Furthermore, disialoganglioside GD2 detection increased the complexity of the expression patterns, giving rise to seven identifiable cell phenotypes. Variation of standard culture conditions, such as the number of cell passage and period in culture, as well as donors, did not influence the heterologous ganglioside expression profile. In contrast, the binding of various lectins appeared homogeneous throughout the MSC population, indicating that the general glycosylation pattern remained common. Morphologically, the expression of a given ganglioside-based phenotype was not related to a cell with particular size or shape. Interestingly, a segregation of GM1 and GM3 clusters was observed, GM3 being mostly excluded from the highly curved plasma membrane protrusions. CONCLUSIONS These data highlight the phenotypic heterogeneity of plastic-adherent MSC in terms of certain lipid constituents of the plasma membrane, and the presence and/or absence of distinct ganglioside-based membrane microdomains suggest their potential functional diversity.
Collapse
Affiliation(s)
- Daniel Freund
- Tissue Engineering Laboratories, BIOTEC and DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | | | | | | | | |
Collapse
|
20
|
Jaime-Pérez JC, Guillermo-Villanueva VA, Méndez-Ramírez N, Chapa-Rodríguez A, Gutiérrez-Aguirre H, Gómez-Almaguer D. CD133+ cell content does not influence recovery time after hematopoietic stem cell transplantation. Transfusion 2010; 49:2390-4. [PMID: 19903294 DOI: 10.1111/j.1537-2995.2009.02292.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Infusion of an adequate dose of CD34+ mononuclear hematopoietic stem cells (HSCs) is the single most important variable to assure success in hematopoietic grafting. CD133+ HSCs constitute the CD34+ subgroup with higher differentiation potential. The number of granulocyte-colony-stimulating factor (G-CSF)-mobilized CD133+ HSCs administered during hematopoietic grafting and its relationship with the number of days needed to regain hematopoiesis was determined. STUDY DESIGN AND METHODS Thirty-eight patients with malignant hematologic diseases who received an autologous (n = 15) or allogeneic (n = 23) HSC transplant were prospectively evaluated. G-CSF was administered for 5 days at 10 microg/kg/day. Hematopoietic progenitors were recovered from peripheral blood on day 5 by leukopheresis. CD34+ and CD133+/CD34+ cell populations were quantified by flow cytometry; the number of days to hematologic recovery was documented. RESULTS A median dose of 4.56 x 10(6)/kg CD34+ HSCs (range, 1.35 x 10(6)-14.6 x 10(6)) was recovered and transplanted; of these grafted cells, a median 3.25 x 10(6) were also CD133+ (range, 1.25 x 10(6)-14.3 x 10(6)). In the autologous group, the median number of days to reach a platelet (PLT) count of 20 x 10(9)/L or greater was 12, and 15 days to obtain a neutrophil count of 0.5 x 10(9)/L or greater; in the allogeneic group 13 and 16 days, respectively, were required (p > 0.05). A median 76.5% of G-CSF-mobilized CD34+ HSCs coexpressed the CD133+ antigen (range, 23.1-97.9). CONCLUSIONS A higher number of CD133+/CD34+ HSCs in the graft was not clearly associated with a shorter neutrophil or PLT recovery time in either allogeneic or autologous recipients.
Collapse
Affiliation(s)
- José Carlos Jaime-Pérez
- Servicio de Hematología, Hospital Universitario Dr José E. González, Facultad de Medicina de la Universidad Autónoma Nuevo León, Monterrey, NL, México.
| | | | | | | | | | | |
Collapse
|
21
|
Raghunath J, Sutherland J, Salih V, Mordan N, Butler PE, Seifalian AM. Chondrogenic potential of blood-acquired mesenchymal progenitor cells. J Plast Reconstr Aesthet Surg 2010; 63:841-7. [DOI: 10.1016/j.bjps.2009.01.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 01/22/2009] [Accepted: 01/30/2009] [Indexed: 12/01/2022]
|
22
|
|
23
|
Jin Y, Lu Z, Ding K, Li J, Du X, Chen C, Sun X, Wu Y, Zhou J, Pan J. Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species. Cancer Res 2010; 70:2516-27. [PMID: 20215516 DOI: 10.1158/0008-5472.can-09-3950] [Citation(s) in RCA: 252] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NF-kappaB may be a potential therapeutic target for acute myelogenous leukemia (AML) because NF-kappaB activation is found in primitive human AML blast cells. In this report, we initially discovered that the potent antineoplastic effect of niclosamide, a Food and Drug Administration-approved antihelminthic agent, was through inhibition of the NF-kappaB pathway in AML cells. Niclosamide inhibited the transcription and DNA binding of NF-kappaB. It blocked tumor necrosis factor-induced IkappaBalpha phosphorylation, translocation of p65, and expression of NF-kappaB-regulated genes. Niclosamide inhibited the steps TAK1-->IkappaB kinase (IKK) and IKK-->IkappaBalpha. Niclosamide also increased the levels of reactive oxygen species (ROS) in AML cells. Quenching ROS by the glutathione precursor N-acetylcysteine attenuated niclosamide-induced apoptosis. Our results together suggest that niclosamide inhibited the NF-kappaB pathway and increased ROS levels to induce apoptosis in AML cells. On translational study of the efficacy of niclosamide against AML, niclosamide killed progenitor/stem cells from AML patients but spared those from normal bone marrow. Niclosamide was synergistic with the frontline chemotherapeutic agents cytarabine, etoposide, and daunorubicin. It potently inhibited the growth of AML cells in vitro and in nude mice. Our results support further investigation of niclosamide in clinical trials of AML patients.
Collapse
Affiliation(s)
- Yanli Jin
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pozzobon M, Piccoli M, Ditadi A, Bollini S, Destro R, André-Schmutz I, Masiero L, Lenzini E, Zanesco L, Petrelli L, Cavazzana-Calvo M, Gazzola MV, De Coppi P. Mesenchymal stromal cells can be derived from bone marrow CD133+ cells: implications for therapy. Stem Cells Dev 2009; 18:497-510. [PMID: 18598159 DOI: 10.1089/scd.2008.0003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
It is known that the bone marrow (BM) CD133(+) cells play an important role in the hematopoietic compartment, but this is not their only role. The cells indeed can take part in vascular reconstitution when they become endothelial cells (EC), in skeletal muscle fiber regeneration when there is a switch in muscle precursors, and to cardiomyocyte phenotypic conversion when differentiating in cardiomyocytes-like cells. While the role in hematopoiesis and vasculogenesis of the selected cells is well established, their ability to differentiate along multiple non-EC lineages has not yet been fully elucidated. The goal of this study is to assert whether human CD133(+)BM-derived cells are able to differentiate in vitro, besides to blood cells, cell lineages pertinent to the mesoderm germ layers. To this end, we isolated CD133(+) cells using a clinically approved methodology and compared their differentiation potential to that of hematopoietic progenitor cells (HPCs) and mesenchymal stem cells (MSCs) obtained from the same BM samples. In our culture conditions, CD133 expression was consistently decreased after passage 2, as well as the expression of the stemness markers c-kit and OCT4, whereas expression of Stage Specific Embryonic Antigen 4 (SSEA4) remained consistent in all different conditions. Expanded CD133 were also positive for HLA-ABC, but negative for HLA-DR, in accordance with what has been previously reported for MSCs. Moreover, CD133(+) cells from human BM demonstrated a wide range of differentiation potential, encompassing not only mesodermal but also ectodermal (neurogenic) cell lineages. CD133 antigen could be potentially used to select a cell population with similar characteristics as MSCs for therapeutic applications.
Collapse
Affiliation(s)
- Michela Pozzobon
- Stem Cell Processing Laboratory and Cord Blood Bank, Department of Pediatric Oncohematology, University of Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Corbeil D, Joester A, Fargeas CA, Jászai J, Garwood J, Hellwig A, Werner HB, Huttner WB. Expression of distinct splice variants of the stem cell marker prominin-1 (CD133) in glial cells. Glia 2009; 57:860-74. [DOI: 10.1002/glia.20812] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
26
|
Kuhara M, Yoshino T, Shiokawa M, Okabe T, Mizoguchi S, Yabuhara A, Takeyama H, Matsunaga T. Magnetic Separation of Human Podocalyxin-like Protein 1 (hPCLP1)-Positive Cells from Peripheral Blood and Umbilical Cord Blood Using Anti-hPCLP1 Monoclonal Antibody and Protein A Expressed on Bacterial Magnetic Particles. Cell Struct Funct 2009; 34:23-30. [DOI: 10.1247/csf.08043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Motoki Kuhara
- Technology and Development Division, Medical & Biological Laboratories Co. Ltd
| | - Tomoko Yoshino
- Department of Biotechnology, Tokyo University of Agriculture and Technology
| | - Miho Shiokawa
- Technology and Development Division, Medical & Biological Laboratories Co. Ltd
| | | | - Shinji Mizoguchi
- Technology and Development Division, Medical & Biological Laboratories Co. Ltd
| | | | - Haruko Takeyama
- Department of Biotechnology, Tokyo University of Agriculture and Technology
| | - Tadashi Matsunaga
- Department of Biotechnology, Tokyo University of Agriculture and Technology
| |
Collapse
|
27
|
Bosio A, Huppert V, Donath S, Hennemann P, Malchow M, Heinlein UAO. Isolation and enrichment of stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2009; 114:23-72. [PMID: 19347268 DOI: 10.1007/10_2008_38] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Stem cells have the potential to revolutionize tissue regeneration and engineering. Both general types of stem cells, those with pluripotent differentiation potential as well as those with multipotent differentiation potential, are of equal interest. They are important tools to further understanding of general cellular processes, to refine industrial applications for drug target discovery and predictive toxicology, and to gain more insights into their potential for tissue regeneration. This chapter provides an overview of existing sorting technologies and protocols, outlines the phenotypic characteristics of a number of different stem cells, and summarizes their potential clinical applications.
Collapse
Affiliation(s)
- Andreas Bosio
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429, Bergisch Gladbach, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Bone marrow-derived stem/progenitor cells: their use in clinical studies for the treatment of myocardial infarction. Heart Lung Circ 2008; 18:171-80. [PMID: 19081302 DOI: 10.1016/j.hlc.2008.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 09/09/2008] [Accepted: 09/10/2008] [Indexed: 01/08/2023]
Abstract
Over the last six years, several centres around the world have started clinical trials to investigate the utilisation of bone marrow-derived cells for myocardial infarction. Different types and numbers of cells have been used assuming they possess a potential to originate new endothelial cells and/or cardiomyocytes to repair/regenerate the ailed heart. Despite diversity in number, clinical status of subjects, route of cell administration, and criteria to evaluate efficacy, the main conclusion drawn from these clinical studies was that such therapies were safe. However, attempts to unify efficacy data have yielded no clear answers, so far. This review offers an in-depth and critical analysis of these trials and intends to evaluate from the cellular biology and clinical cardiology viewpoints, the significant information that has been published since 2002, as well as that emerging from ongoing clinical trials. Emphasis will be placed on cellular types, research designs and methods to evaluate efficacy of each particular treatment modality.
Collapse
|
29
|
Karbanová J, Missol-Kolka E, Fonseca AV, Lorra C, Janich P, Hollerová H, Jászai J, Ehrmann J, Kolár Z, Liebers C, Arl S, Subrtová D, Freund D, Mokry J, Huttner WB, Corbeil D. The stem cell marker CD133 (Prominin-1) is expressed in various human glandular epithelia. J Histochem Cytochem 2008; 56:977-93. [PMID: 18645205 DOI: 10.1369/jhc.2008.951897] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Human prominin-1 (CD133) is expressed by various stem and progenitor cells originating from diverse sources. In addition to stem cells, its mouse ortholog is expressed in a broad range of adult epithelial cells, where it is selectively concentrated in their apical domain. The lack of detection of prominin-1 in adult human epithelia might be explained, at least in part, by the specificity of the widely used AC133 antibody, which recognizes an epitope that seems dependent on glycosylation. Here we decided to re-examine its expression in adult human tissues, particularly in glandular epithelia, using a novel monoclonal antibody (80B258) generated against the human prominin-1 polypeptide. In examined tissues, we observed 80B258 immunoreactivity at the apical or apicolateral membranes of polarized cells. For instance, we found expression in secretory serous and mucous cells as well as intercalated ducts of the large salivary and lacrimal glands. In sweat glands including the gland of Moll, 80B258 immunoreactivity was found in the secretory (eccrine and apocrine glands) and duct (eccrine glands) portion. In the liver, 80B258 immunoreactivity was identified in the canals of Hering, bile ductules, and small interlobular bile ducts. In the uterus, we detected 80B258 immunoreactivity in endometrial and cervical glands. Together these data show that the overall expression of human prominin-1 is beyond the rare primitive cells, and it seems to be a general marker of apical or apicolateral membrane of glandular epithelia. This manuscript contains online supplemental material at http://www.jhc.org. Please visit this article online to view these materials.
Collapse
Affiliation(s)
- Jana Karbanová
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Hradec Králové, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bauer N, Fonseca AV, Florek M, Freund D, Jászai J, Bornhäuser M, Fargeas CA, Corbeil D. New insights into the cell biology of hematopoietic progenitors by studying prominin-1 (CD133). Cells Tissues Organs 2007; 188:127-38. [PMID: 18160824 DOI: 10.1159/000112847] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Prominin-1 (alias CD133) has received considerable interest because of its expression by several stem and progenitor cells originating from various sources, including the neural and hematopoietic systems. As a cell surface marker, prominin-1 is now used for somatic stem cell isolation. Its expression in cancer stem cells has broadened its clinical value, as it might be useful to outline new prospects for more effective cancer therapies by targeting tumor-initiating cells. Cell biological studies of this molecule have demonstrated that it is specifically concentrated in various membrane structures that protrude from the planar areas of the plasmalemma. Prominin-1 binds to the plasma membrane cholesterol and is associated with a particular membrane microdomain in a cholesterol-dependent manner. Although its physiological function is not yet determined, it is becoming clear that this cell surface protein, as a unique marker of both plasma membrane protrusions and membrane microdomains, might reveal new aspects of the cell biology of rare stem and cancer stem cells. The aim of this review is to outline the recent discoveries regarding the dynamic reorganization of the plasma membrane of rare CD133+ hematopoietic progenitor cells during cell migration and division.
Collapse
Affiliation(s)
- Nicola Bauer
- Tissue Engineering Laboratories, Biotec, Technische Universität Dresden, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kamprad M, Kindler S, Schuetze N, Emmrich F. Flow Cytometric Immunophenotyping of Umbilical Cord and Peripheral Blood Haematopoietic Progenitor Cells by Different CD34 Epitopes, CD133, P-Glycoprotein Expression and Rhodamine-123 Efflux. Transfus Med Hemother 2007. [DOI: 10.1159/000101555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
32
|
Freund D, Bauer N, Boxberger S, Feldmann S, Streller U, Ehninger G, Werner C, Bornhäuser M, Oswald J, Corbeil D. Polarization of human hematopoietic progenitors during contact with multipotent mesenchymal stromal cells: effects on proliferation and clonogenicity. Stem Cells Dev 2007; 15:815-29. [PMID: 17253945 DOI: 10.1089/scd.2006.15.815] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Establishment of a defined cell culture system that facilitates ex vivo expansion of isolated hematopoietic stem and progenitor cells (HSPCs) is a crucial issue in hematology and stem cell transplantation. Here we have evaluated the capacity of primary human multipotent mesenchymal stromal cells (MSCs) to support the ex vivo expansion of peripheral CD34(+)-enriched HSPCs. We observed that HSPCs co-cultured on MSCs showed a substantially higher total expansion rate compared to those growing without. Moreover, in addition to the expansion of CD34(+)CD133(+) and CD34(+)CD133(-) cells, a third population of CD133(+)CD34(-) stem cells became detectable after expansion. Direct contact between HSPCs and the feeder layer appears beneficial for the expansion of HSPCs harboring CD133(+) phenotype, i.e., CD34(+)CD133(+) and CD133(+)CD34(-), in contrast to CD34(+)CD133(-) cells. Interestingly, electron microscopy and immunofluorescence analyses revealed that adherent HSPCs display various morphologies; they are either round with, in some cases, the appearance of a microvillar pole or exhibit several distinct types of plasma membrane protrusions such as lamellipodium and magnupodium. CD133 is selectively concentrated therein, whereas CD34 is randomly distributed over the entire surface of HSPCs. Together, this co-culture offers a unique experimental system to further characterize the biology and role of markers of rare stem cell populations.
Collapse
Affiliation(s)
- Daniel Freund
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, D-01307, Dresden, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lu X, Baudouin SV, Gillespie JI, Anderson JJ, Dickinson AM. A comparison of CFU-GM, BFU-E and endothelial progenitor cells using ex vivo expansion of selected cord blood CD133+ and CD34+ cells. Cytotherapy 2007; 9:292-300. [PMID: 17464761 DOI: 10.1080/14653240701247853] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND CD133 is a newly developed hematopoietic stem cell marker but little is known about its function. Whether CD133(+) cell selection provides any advantage over CD34(+) selection for hematopoietic stem cell isolation and transplantation is unclear. The present study compared colony formation and endothelial cell differentiation of these two cell types from umbilical cord blood (UCB). METHODS Mononuclear cells from the same UCB samples were used for both CD133(+) and CD34(+) cell selection. Cells with 97.1% purity were incubated in semi-solid culture medium containing stem cell growth factor (SCGF) and G-CSF or erythropoietin (EPO). Purified cells were also cultured in M199 containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and insulin-like growth factor-1 (IGF-1). RESULTS CD34(+) and CD133(+) cells produced similar numbers of CFU-GM colonies (median 43.25 and 30.5, respectively; P>0.2). However, a greater than four-fold difference in BFU-E colony formation was observed from CD34(+) cells compared with CD133(+) cells (median 35 and 8, respectively; P<0.04). CD34(+) cells gave rise to endothelial-like cells when stimulated with VEGF, bFGF and IGF-1. CD133(+) cells were unable produce this cell type under the same conditions. DISCUSSION CD133(+) cells produced smaller BFU-E colonies and were unable to differentiate into mature endothelial cells. CD34(+) cells contained endothelial progenitors that could differentiate into mature cells of this lineage. Based on these data, it appears that CD133 offers no distinct advantage over CD34 as a selective marker for immunoaffinity-based isolation of hematopoietic stem cells and endothelial progenitor cells.
Collapse
Affiliation(s)
- X Lu
- Haematological Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, NE2 4HH, UK.
| | | | | | | | | |
Collapse
|