1
|
Lissek T. Enhancement of physiology via adaptive transcription. Pflugers Arch 2025; 477:187-199. [PMID: 39482558 PMCID: PMC11761519 DOI: 10.1007/s00424-024-03037-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
The enhancement of complex physiological functions such as cognition and exercise performance in healthy individuals represents a challenging goal. Adaptive transcription programs that are naturally activated in animals to mediate cellular plasticity in response to stimulation can be leveraged to enhance physiological function above wild-type levels in young organisms and counteract complex functional decline in aging. In processes such as learning and memory and exercise-dependent muscle remodeling, a relatively small number of molecules such as certain stimulus-responsive transcription factors and immediate early genes coordinate widespread changes in cellular physiology. Adaptive transcription can be targeted by various methods including pharmaceutical compounds and gene transfer technologies. Important problems for leveraging adaptive transcription programs for physiological enhancement include a better understanding of their dynamical organization, more precise methods to influence the underlying molecular components, and the integration of adaptive transcription into multi-scale physiological enhancement concepts.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Balmas E, Sozza F, Bottini S, Ratto ML, Savorè G, Becca S, Snijders KE, Bertero A. Manipulating and studying gene function in human pluripotent stem cell models. FEBS Lett 2023; 597:2250-2287. [PMID: 37519013 DOI: 10.1002/1873-3468.14709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023]
Abstract
Human pluripotent stem cells (hPSCs) are uniquely suited to study human development and disease and promise to revolutionize regenerative medicine. These applications rely on robust methods to manipulate gene function in hPSC models. This comprehensive review aims to both empower scientists approaching the field and update experienced stem cell biologists. We begin by highlighting challenges with manipulating gene expression in hPSCs and their differentiated derivatives, and relevant solutions (transfection, transduction, transposition, and genomic safe harbor editing). We then outline how to perform robust constitutive or inducible loss-, gain-, and change-of-function experiments in hPSCs models, both using historical methods (RNA interference, transgenesis, and homologous recombination) and modern programmable nucleases (particularly CRISPR/Cas9 and its derivatives, i.e., CRISPR interference, activation, base editing, and prime editing). We further describe extension of these approaches for arrayed or pooled functional studies, including emerging single-cell genomic methods, and the related design and analytical bioinformatic tools. Finally, we suggest some directions for future advancements in all of these areas. Mastering the combination of these transformative technologies will empower unprecedented advances in human biology and medicine.
Collapse
Affiliation(s)
- Elisa Balmas
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Federica Sozza
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Sveva Bottini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Maria Luisa Ratto
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Giulia Savorè
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Silvia Becca
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Kirsten Esmee Snijders
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Turin, Torino, Italy
| |
Collapse
|
3
|
Wu M, Liu S, Gao Y, Bai H, Machairaki V, Li G, Chen T, Cheng L. Conditional gene knockout and reconstitution in human iPSCs with an inducible Cas9 system. Stem Cell Res 2018; 29:6-14. [DOI: 10.1016/j.scr.2018.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 12/11/2022] Open
|
4
|
Gabner S, Hlavaty J, Velde K, Renner M, Jenner F, Egerbacher M. Inflammation-induced transgene expression in genetically engineered equine mesenchymal stem cells. J Gene Med 2018; 18:154-64. [PMID: 27272202 DOI: 10.1002/jgm.2888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 05/31/2016] [Accepted: 05/31/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Osteoarthritis, a chronic and progressive degenerative joint disorder, ranks amongst the top five causes of disability. Given the high incidence, associated socioeconomic costs and the absence of effective disease-modifying therapies of osteoarthritis, cell-based treatments offer a promising new approach. Owing to their paracrine, differentiation and self-renewal abilities, mesenchymal stem cells (MSCs) have great potential for regenerative medicine, which might be further enhanced by targeted gene therapy. Hence, the development of systems allowing transgene expression, particularly when regulated by natural disease-dependent occuring substances, is of high interest. METHODS Bone marrow-isolated equine MSCs were stably transduced with an HIV-1 based lentiviral vector expressing the luciferase gene under control of an inducible nuclear factor κB (NFκB)-responsive promoter. Marker gene expression was analysed by determining luciferase activity in transduced cells stimulated with different concentrations of interleukin (IL)-1β or tumour necrosis factor (TNF)α. RESULTS A dose-dependent increase in luciferase expression was observed in transduced MSCs upon cytokine stimulation. The induction effect was more potent in cells treated with TNFα compared to those treated with IL-1β. Maximum transgene expression was obtained after 48 h of stimulation and the same time was necessary to return to baseline luciferase expression levels after withdrawal of the stimulus. Repeated cycles of induction allowed on-off modulation of transgene expression without becoming refractory to induction. The NFκB-responsive promoter retained its inducibility also in chondrogenically differentiated MSC/Luc cells. CONCLUSIONS The results of the present study demonstrate that on demand transgene expression from the NFκB-responsive promoter using naturally occurring inflammatory cytokines can be induced in undifferentiated and chondrogenically differentiated equine MSCs. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Simone Gabner
- Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Juraj Hlavaty
- Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karsten Velde
- Equine University Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Matthias Renner
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, Langen, Germany
| | - Florien Jenner
- Equine University Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Monika Egerbacher
- Institute of Anatomy, Histology and Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
5
|
Wu RA, Tam J, Collins K. DNA-binding determinants and cellular thresholds for human telomerase repeat addition processivity. EMBO J 2017; 36:1908-1927. [PMID: 28495680 PMCID: PMC5494469 DOI: 10.15252/embj.201796887] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 12/30/2022] Open
Abstract
The reverse transcriptase telomerase adds telomeric repeats to chromosome ends. Purified human telomerase catalyzes processive repeat synthesis, which could restore the full ~100 nucleotides of (T2AG3)n lost from replicated chromosome ends as a single elongation event. Processivity inhibition is proposed to be a basis of human disease, but the impacts of different levels of processivity on telomere maintenance have not been examined. Here, we delineate side chains in the telomerase active-site cavity important for repeat addition processivity, determine how they contribute to duplex and single-stranded DNA handling, and test the cellular consequences of partial or complete loss of repeat addition processivity for telomere maintenance. Biochemical findings oblige a new model for DNA and RNA handling dynamics in processive repeat synthesis. Biological analyses implicate repeat addition processivity as essential for telomerase function. However, telomeres can be maintained by telomerases with lower than wild-type processivity. Furthermore, telomerases with low processivity dramatically elongate telomeres when overexpressed. These studies reveal distinct consequences of changes in telomerase repeat addition processivity and expression level on telomere elongation and length maintenance.
Collapse
Affiliation(s)
- Robert Alexander Wu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jane Tam
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kathleen Collins
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
6
|
Lent-On-Plus Lentiviral vectors for conditional expression in human stem cells. Sci Rep 2016; 6:37289. [PMID: 27853296 PMCID: PMC5112523 DOI: 10.1038/srep37289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 10/28/2016] [Indexed: 12/25/2022] Open
Abstract
Conditional transgene expression in human stem cells has been difficult to achieve due to the low efficiency of existing delivery methods, the strong silencing of the transgenes and the toxicity of the regulators. Most of the existing technologies are based on stem cells clones expressing appropriate levels of tTA or rtTA transactivators (based on the TetR-VP16 chimeras). In the present study, we aim the generation of Tet-On all-in-one lentiviral vectors (LVs) that tightly regulate transgene expression in human stem cells using the original TetR repressor. By using appropriate promoter combinations and shielding the LVs with the Is2 insulator, we have constructed the Lent-On-Plus Tet-On system that achieved efficient transgene regulation in human multipotent and pluripotent stem cells. The generation of inducible stem cell lines with the Lent-ON-Plus LVs did not require selection or cloning, and transgene regulation was maintained after long-term cultured and upon differentiation toward different lineages. To our knowledge, Lent-On-Plus is the first all-in-one vector system that tightly regulates transgene expression in bulk populations of human pluripotent stem cells and its progeny.
Collapse
|
7
|
Li P, Li M, Tang X, Wang S, Zhang YA, Chen Z. Accelerated generation of oligodendrocyte progenitor cells from human induced pluripotent stem cells by forced expression of Sox10 and Olig2. SCIENCE CHINA-LIFE SCIENCES 2016; 59:1131-1138. [PMID: 27785726 DOI: 10.1007/s11427-016-0165-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/31/2016] [Indexed: 10/20/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) hold great promise for treatment of dysmyelinating disorders, such as multiple sclerosis and cerebral palsy. Recent studies on generation of human OPCs mainly use human embryonic stem cells (hESCs) or neural stem cells (NSCs) as starter cell sources for the differentiation process. However, NSCs are restricted in availability and the present method for generation of oligodendrocytes (OLs) from ESCs often requires a lengthy period of time. Here, we demonstrated a protocol to efficiently derive OPCs from human induced pluripotent stem cells (hiPSCs) by forced expression of two transcription factors (2TFs), Sox10 and Olig2. With this method, PDGFRα+ OPCs can be obtained in 14 days and O4+ OPCs in 56 days. Furthermore, OPCs may be able to differentiate to mature OLs that could ensheath axons when co-cultured with rat cortical neurons. The results have implications in the development of autologous cell therapies.
Collapse
Affiliation(s)
- Pengyan Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, and Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China.,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mo Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, and Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Xihe Tang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, and Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Shuyan Wang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, and Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Y Alex Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, and Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China. .,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China.
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, and Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China. .,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China. .,Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
8
|
Vogan JM, Zhang X, Youmans DT, Regalado SG, Johnson JZ, Hockemeyer D, Collins K. Minimized human telomerase maintains telomeres and resolves endogenous roles of H/ACA proteins, TCAB1, and Cajal bodies. eLife 2016; 5. [PMID: 27525486 PMCID: PMC5005035 DOI: 10.7554/elife.18221] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/14/2016] [Indexed: 01/22/2023] Open
Abstract
We dissected the importance of human telomerase biogenesis and trafficking pathways for telomere maintenance. Biological stability of human telomerase RNA (hTR) relies on H/ACA proteins, but other eukaryotes use other RNP assembly pathways. To investigate additional rationale for human telomerase assembly as H/ACA RNP, we developed a minimized cellular hTR. Remarkably, with only binding sites for telomerase reverse transcriptase (TERT), minimized hTR assembled biologically active enzyme. TERT overexpression was required for cellular interaction with minimized hTR, indicating that H/ACA RNP assembly enhances endogenous hTR-TERT interaction. Telomere maintenance by minimized telomerase was unaffected by the elimination of the telomerase holoenzyme Cajal body chaperone TCAB1 or the Cajal body scaffold protein Coilin. Surprisingly, wild-type hTR also maintained and elongated telomeres in TCAB1 or Coilin knockout cells, with distinct changes in telomerase action. Overall, we elucidate trafficking requirements for telomerase biogenesis and function and expand mechanisms by which altered telomere maintenance engenders human disease. DOI:http://dx.doi.org/10.7554/eLife.18221.001 Most cells in the human body can only divide a certain number of times before they die. This is because regions called telomeres at the ends of the cell’s DNA get shorter every time the cell divides, to the point that they disappear and halt cell growth. Particular types of cells – including some stem cells and cancer cells – can avoid death and continue to divide indefinitely because they produce an enzyme called telomerase that extends the telomere regions. The process by which the telomerase enzyme binds to and lengthens the DNA has several stages and involves many different proteins. One of the stages involves moving telomerase from the sites where it is assembled within the cell to a place where it can find telomeres in need of elongation (different areas within the cell compartment called the nucleus). Structures inside the nucleus called Cajal bodies were thought to help the enzyme bind to the telomeres. It is not clear why the process of extending telomeres is so complex. Vogan et al. engineered altered versions of telomerase that use simpler pathways to bind to and act on telomeres and inserted them into ‘pluripotent’ stem cells and cancer cells from humans. The experiments show that a pathway that helps to move the enzyme from its normal storage place in the nucleus is less important for extending telomeres in cancer cells than in pluripotent stem cells. Unexpectedly, Cajal bodies are not critical for bringing telomerase into contact with the telomeres in either cell type. The findings show that many of the proteins involved in extending telomeres in cells are not strictly essential. The simplified pathway developed by Vogan et al. opens up new opportunities to study the details of how telomerase extends telomeres. DOI:http://dx.doi.org/10.7554/eLife.18221.002
Collapse
Affiliation(s)
- Jacob M Vogan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Xiaozhu Zhang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Daniel T Youmans
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Samuel G Regalado
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Joshua Z Johnson
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Kathleen Collins
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
9
|
Wang F, Okawa H, Kamano Y, Niibe K, Kayashima H, Osathanon T, Pavasant P, Saeki M, Yatani H, Egusa H. Controlled Osteogenic Differentiation of Mouse Mesenchymal Stem Cells by Tetracycline-Controlled Transcriptional Activation of Amelogenin. PLoS One 2015; 10:e0145677. [PMID: 26709694 PMCID: PMC4692545 DOI: 10.1371/journal.pone.0145677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/07/2015] [Indexed: 01/25/2023] Open
Abstract
Regenerative dental therapies for bone tissues rely on efficient targeting of endogenous and transplanted mesenchymal stem cells (MSCs) to guide bone formation. Amelogenin is the primary component of Emdogain, which is used to regenerate periodontal defects; however, the mechanisms underlying the therapeutic effects on alveolar bone remain unclear. The tetracycline (Tet)-dependent transcriptional regulatory system is a good candidate to investigate distinct roles of genes of interest during stem cell differentiation. Here, we investigated amelogenin-dependent regulation of osteogenesis in MSCs by establishing a Tet-controlled transcriptional activation system. Clonal mouse bone marrow-derived MSCs were lentivirally transduced with the Tet repressor (TetR) expression vector followed by drug selection to obtain MSCs constitutively expressing TetR (MSCs-TetR). Expression vectors that contained the Tet operator and amelogenin-coding (Amelx) cDNA fragments were constructed using the Gateway system and lentivirally introduced into MSCs-TetR to generate a Tet regulation system in MSCs (MSCs-TetR/Amelx). MSCs-TetR/Amelx significantly overexpressed the Amelx gene and protein in the presence of the tetracycline derivative doxycycline. Concomitant expression of osterix, bone sialoprotein (BSP), osteopontin, and osteocalcin was modulated by addition or removal of doxycycline under osteogenic guidance. During osteogenic induction, MSCs-TetR/Amelx treated with doxycycline showed significantly increased gene expression of osterix, type I collagen, BSP, and osteocalcin in addition to increased alkaline phosphatase activity and mineralized nodule formation. Enhanced extracellular matrix calcification was observed when forced Amelx expression commenced at the early stage but not at the intermediate or late stages of osteogenesis. These results suggest that a Tet-controlled Amelx gene regulation system for mouse MSCs was successfully established, in which transcriptional activation of Amelx was associated with enhanced osteogenic differentiation, especially in the early stage of biomineralization.
Collapse
Affiliation(s)
- Fangfang Wang
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hiroko Okawa
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Yuya Kamano
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Kunimichi Niibe
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Hiroki Kayashima
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Thanaphum Osathanon
- Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Research Unit of Mineralized Tissue, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Makio Saeki
- Division of Dental Pharmacology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hirofumi Yatani
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Hiroshi Egusa
- Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
- * E-mail:
| |
Collapse
|
10
|
Lachmann N, Brennig S, Hillje R, Schermeier H, Phaltane R, Dahlmann J, Gruh I, Heinz N, Schiedlmeier B, Baum C, Moritz T. Tightly regulated 'all-in-one' lentiviral vectors for protection of human hematopoietic cells from anticancer chemotherapy. Gene Ther 2015; 22:883-92. [PMID: 26125609 DOI: 10.1038/gt.2015.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 06/09/2015] [Accepted: 06/18/2015] [Indexed: 01/12/2023]
Abstract
Successful application of gene therapy strategies may require stringently regulated transgene expression. Along this line, we describe a doxycycline (Dox)-inducible 'all-in-one' lentiviral vector design using the pTET-T11 (TII) minimal-promoter and a reverse transactivator protein (rtTA2S-M2) driven by the phosphoglycerate kinase promoter allowing for tight regulation of transgene expression (Lv.TII vectors). Vector design was evaluated in human hematopoietic cells in the context of cytidine deaminase (hCDD)-based myeloprotective gene therapy. Upon Dox administration, a rapid (16-24 h) and dose-dependent (>0.04 μg ml(-1) Dox) onset of transgene expression was detected in Lv.TII.CDD gene-modified K562 cells as well as in primary human CD34(+) hematopoietic cells. Importantly, in both cell models low background transgene expression was observed in the absence of Dox. Functionality of Dox-inducible hCDD expression was demonstrated by >10-fold increase in cytosine arabinoside (1-β-d-arabinofuranosylcytosine, Ara-C) resistance of Lv.TII.CDD-transduced K562 cells. In addition, Lv.TII.CDD-transduced CD34(+)-derived myeloid cells were protected from up to 300 nm Ara-C (control affected from 50 nm onwards). These data clearly demonstrate the suitability of our self-inactivating lentiviral vector to induce robust, tightly regulated transgene expression in human hematopoietic cells with minimal background activity and highlight the potential of our construct in myeloprotective gene therapy strategies.
Collapse
Affiliation(s)
- N Lachmann
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - S Brennig
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - R Hillje
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - H Schermeier
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - R Phaltane
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - J Dahlmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - I Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - N Heinz
- LOEWE-Research Group for (targeted) Gene Modification in Stem Cells, Paul-Ehrlich-Institute, Langen, Germany
| | - B Schiedlmeier
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - C Baum
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - T Moritz
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Qian K, Huang CTL, Huang CL, Chen H, Blackbourn LW, Chen Y, Cao J, Yao L, Sauvey C, Du Z, Zhang SC. A simple and efficient system for regulating gene expression in human pluripotent stem cells and derivatives. Stem Cells 2014; 32:1230-8. [PMID: 24497442 DOI: 10.1002/stem.1653] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/06/2014] [Indexed: 12/19/2022]
Abstract
Regulatable transgene expression in human pluripotent stem cells (hPSCs) and their progenies is often necessary to dissect gene function in a temporal and spatial manner. However, hPSC lines with inducible transgene expression, especially in differentiated progenies, have not been established due to silencing of randomly inserted genes during stem cell expansion and/or differentiation. Here, we report the use of transcription activator-like effector nucleases-mediated targeting to AAVS1 site to generate versatile conditional hPSC lines. Transgene (both green fluorescent protein and a functional gene) expression in hPSCs and their derivatives was not only sustained but also tightly regulated in response to doxycycline both in vitro and in vivo. We modified the donor construct so that any gene of interest can be readily inserted to produce hPSC lines with conditional transgene expression. This technology will substantially improve the way we study human stem cells.
Collapse
Affiliation(s)
- Kun Qian
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Waisman Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Intrastriatal GDNF gene transfer by inducible lentivirus vectors protects dopaminergic neurons in a rat model of parkinsonism. Exp Neurol 2014; 261:87-96. [DOI: 10.1016/j.expneurol.2014.06.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/18/2014] [Accepted: 06/20/2014] [Indexed: 11/20/2022]
|
13
|
Human herpesvirus 8 viral interleukin-6 signaling through gp130 promotes virus replication in primary effusion lymphoma and endothelial cells. J Virol 2014; 88:12167-72. [PMID: 25078695 DOI: 10.1128/jvi.01751-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The contributions of human herpesvirus 8 (HHV-8) viral interleukin-6 (vIL-6) to virus biology remain unclear. Here we examined the role of vIL-6/gp130 signaling in HHV-8 productive replication in primary effusion lymphoma and endothelial cells. Depletion and depletion-complementation experiments revealed that endoplasmic reticulum-localized vIL-6 activity via gp130 and gp130-activated signal transducer and activator of transcription (STAT) signaling, but not extracellular signal-regulated kinase (ERK) activation, was critical for vIL-6 proreplication activity. Our data significantly extend current understanding of vIL-6 function and associated mechanisms in HHV-8 biology.
Collapse
|
14
|
Hartman ME, Librande JR, Medvedev IO, Ahmad RN, Moussavi-Harami F, Gupta PP, Chien WM, Chin MT. An optimized and simplified system of mouse embryonic stem cell cardiac differentiation for the assessment of differentiation modifiers. PLoS One 2014; 9:e93033. [PMID: 24667642 PMCID: PMC3965510 DOI: 10.1371/journal.pone.0093033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/28/2014] [Indexed: 12/19/2022] Open
Abstract
Generating cardiomyocytes from embryonic stem cells is an important technique for understanding cardiovascular development, the origins of cardiovascular diseases and also for providing potential reagents for cardiac repair. Numerous methods have been published but often are technically challenging, complex, and are not easily adapted to assessment of specific gene contributions to cardiac myocyte differentiation. Here we report the development of an optimized protocol to induce the differentiation of mouse embryonic stem cells to cardiac myocytes that is simplified and easily adapted for genetic studies. Specifically, we made four critical findings that distinguish our protocol: 1) mouse embryonic stem cells cultured in media containing CHIR99021 and PD0325901 to maintain pluripotency will efficiently form embryoid bodies containing precardiac mesoderm when cultured in these factors at a reduced dosage, 2) low serum conditions promote cardiomyocyte differentiation and can be used in place of commercially prepared StemPro nutrient supplement, 3) the Wnt inhibitor Dkk-1 is dispensable for efficient cardiac differentiation and 4) tracking differentiation efficiency may be done with surface expression of PDGFRα alone. In addition, cardiac mesodermal precursors generated by this system can undergo lentiviral infection to manipulate the expression of specific target molecules to assess effects on cardiac myocyte differentiation and maturation. Using this approach, we assessed the effects of CHF1/Hey2 on cardiac myocyte differentiation, using both gain and loss of function. Overexpression of CHF1/Hey2 at the cardiac mesoderm stage had no apparent effect on cardiac differentiation, while knockdown of CHF1/Hey2 resulted in increased expression of atrial natriuretic factor and connexin 43, suggesting an alteration in the phenotype of the cardiomyocytes. In summary we have generated a detailed and simplified protocol for generating cardiomyocytes from mES cells that is optimized for investigating factors that affect cardiac differentiation.
Collapse
Affiliation(s)
- Matthew E. Hartman
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jason R. Librande
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ivan O. Medvedev
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Rabiah N. Ahmad
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Farid Moussavi-Harami
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Pritha P. Gupta
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Wei-Ming Chien
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Michael T. Chin
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
15
|
Tsai SC, Chang DF, Hong CM, Xia P, Senadheera D, Trump L, Mishra S, Lutzko C. Induced overexpression of OCT4A in human embryonic stem cells increases cloning efficiency. Am J Physiol Cell Physiol 2014; 306:C1108-18. [PMID: 24627557 DOI: 10.1152/ajpcell.00205.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Our knowledge of the molecular mechanisms underlying human embryonic stem cell (hESC) self-renewal and differentiation is incomplete. The level of octamer-binding transcription factor 4 (Oct4), a critical regulator of pluripotency, is precisely controlled in mouse embryonic stem cells. However, studies of human OCT4 are often confounded by the presence of three isoforms and six expressed pseudogenes, which has complicated the interpretation of results. Using an inducible lentiviral overexpression and knockdown system to manipulate OCT4A above or below physiological levels, we specifically examine the functional role of the OCT4A isoform in hESC. (We also designed and generated a comparable series of vectors, which were not functional, for the overexpression and knockdown of OCT4B.) We show that specific knockdown of OCT4A results in hESC differentiation, as indicated by morphology changes, cell surface antigen expression, and upregulation of ectodermal genes. In contrast, inducible overexpression of OCT4A in hESC leads to a transient instability of the hESC phenotype, as indicated by changes in morphology, cell surface antigen expression, and transcriptional profile, that returns to baseline within 5 days. Interestingly, sustained expression of OCT4A past 5 days enhances hESC cloning efficiency, suggesting that higher levels of OCT4A can support self-renewal. Overall, our results indicate that high levels of OCT4A increase hESC cloning efficiency and do not induce differentiation (whereas OCT4B expression cannot be induced in hESC), highlighting the importance of isoform-specific studies in a stable and inducible expression system for human OCT4. Additionally, we demonstrate the utility of an efficient method for conditional gene expression in hESC.
Collapse
Affiliation(s)
- Steven C Tsai
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - David F Chang
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Chang-Mu Hong
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Ping Xia
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Dinithi Senadheera
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Lisa Trump
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Children's Hospital Research Foundation, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - Suparna Mishra
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Carolyn Lutzko
- Division of Research Immunology and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, California; Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Children's Hospital Research Foundation, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and Division of Regenerative Medicine and Cellular Therapies, Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
16
|
Human herpesvirus 8 interleukin-6 contributes to primary effusion lymphoma cell viability via suppression of proapoptotic cathepsin D, a cointeraction partner of vitamin K epoxide reductase complex subunit 1 variant 2. J Virol 2013; 88:1025-38. [PMID: 24198402 DOI: 10.1128/jvi.02830-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human herpesvirus 8 (HHV-8) interleukin-6 (vIL-6) promotes cell proliferation and survival and is proangiogenic, implicating it as a contributor to virus-associated Kaposi's sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman's disease. Although predominantly lytically expressed, vIL-6 is also produced at low, functional levels during latency in PEL cells. Unlike other IL-6 cytokines, vIL-6 is secreted very inefficiently and localizes in the endoplasmic reticulum (ER). ER-localized vIL-6 supports PEL cell proliferation and survival, mediated in part through its interaction with the largely uncharacterized ER-resident protein vitamin K epoxide reductase complex subunit 1 variant 2 (VKORC1v2). Here, we report that the ER-transiting and functionally mitogenic secreted proenzyme (pCatD) form of cathepsin D (mature CatD), a proapoptotic lysosomal aspartate protease, is an interaction partner of VKORC1v2 and that vIL-6 promotes this interaction. Depletion of vIL-6 in PEL cells increased levels of the catalytically active, proteolytically cleaved form of CatD, corresponding with decreased PEL cell viability. Ectopic expression of CatD in PEL cells induced apoptosis, suggesting that CatD suppression by vIL-6 is biologically significant. In the context of high-density culture or reactivation of HHV-8 lytic replication in PEL cells, CatD depletion substantially reduced stress-induced apoptosis and increased virus production. In contrast, CatD overexpression, vIL-6 depletion, and peptide-mediated disruption of vIL-6-VKORC1v2 interaction inhibited replication and cell survival. Combined, our data identify pCatD as an interaction partner of VKORC1v2, demonstrate a role of vIL-6 in CatD suppression via VKORC1v2 in PEL cells, and identify a biologically significant mechanism of vIL-6 prosurvival and proreplication activities via VKORC1v2.
Collapse
|
17
|
Yuan X, Braunstein EM, Ye Z, Liu CF, Chen G, Zou J, Cheng L, Brodsky RA. Generation of glycosylphosphatidylinositol anchor protein-deficient blood cells from human induced pluripotent stem cells. Stem Cells Transl Med 2013; 2:819-29. [PMID: 24113066 DOI: 10.5966/sctm.2013-0069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
PIG-A is an X-linked gene required for the biosynthesis of glycosylphosphatidylinositol (GPI) anchors; thus, PIG-A mutant cells have a deficiency or absence of all GPI-anchored proteins (GPI-APs). Acquired mutations in hematopoietic stem cells result in the disease paroxysmal nocturnal hemoglobinuria, and hypomorphic germline PIG-A mutations lead to severe developmental abnormalities, seizures, and early death. Human induced pluripotent stem cells (iPSCs) can differentiate into cell types derived from all three germ layers, providing a novel developmental system for modeling human diseases. Using PIG-A gene targeting and an inducible PIG-A expression system, we have established, for the first time, a conditional PIG-A knockout model in human iPSCs that allows for the production of GPI-AP-deficient blood cells. PIG-A-null iPSCs were unable to generate hematopoietic cells or any cells expressing the CD34 marker and were defective in generating mesodermal cells expressing KDR/VEGFR2 (kinase insert domain receptor) and CD56 markers. In addition, PIG-A-null iPSCs had a block in embryonic development prior to mesoderm differentiation that appears to be due to defective signaling through bone morphogenetic protein 4. However, early inducible PIG-A transgene expression allowed for the generation of GPI-AP-deficient blood cells. This conditional PIG-A knockout model should be a valuable tool for studying the importance of GPI-APs in hematopoiesis and human development.
Collapse
Affiliation(s)
- Xuan Yuan
- Division of Hematology, Department of Medicine, School of Medicine, and
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Guan Y, Zou H, Chen X, Zhao C, Wang J, Cai Y, Chan P, Chen L, Zhang YA. Ischemia, immunosuppression, and SSEA-1-negative cells all contribute to tumors resulting from mouse embryonic stem cell-derived neural progenitor transplantation. J Neurosci Res 2013; 92:74-85. [PMID: 24123213 DOI: 10.1002/jnr.23292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 08/06/2013] [Accepted: 08/08/2013] [Indexed: 01/16/2023]
Abstract
Neural progenitor cells (NPCs) derived from mouse embryonic stem (mES) cells can lead to tumors after transplantation. The cellular source of such tumors remains under debate. We investigated the tumor formation resulting from mES cell-derived NPCs in a rat stroke model and in nude mice. After 2 hr of ischemia and 48 hr of reperfusion, the NPCs were transplanted into the ischemic core of the xenogeneic rats. Four weeks after transplantation, the grafted cells were found to be viable at the border of the necrosis and had differentiated into neurons. Transplanted rats did not exhibit any behavioral improvement, because tumor formed in 90% of the animals. Immunosuppression facilitated tumor formation. Tumors were observed in 40% of normal rats after NPC transplantation when cyclosporin A was administered. Meanwhile, no tumor formation was observed without cyclosporin A. Ischemic damage also facilitated tumor formation, because NPCs gave rise to tumors in 90% of ischemic rats, a percentage significantly higher than that in intact rats, which was 40%. The SSEA-1-positive cells isolated from stage 4 are not exactly undifferentiated ES cells. They exhibited a marker gene transcription profile different from that of ES cells and did not form tumors in transplanted nude mice. The undifferentiated ES cells remaining after differentiation did not contribute to tumors either. First, the tumor formation rate resulting from undifferentiated ES cells in the brains of normal rats is 0%, significantly lower than that of NPCs. Second, transplanted NPCs that led to 100% tumors in nude mice contained approximately 1.5 × 10(3) Oct-4-positive cells; however, even 5 × 10(5) undifferentiated ES cells formed neoplasm only in 40% nude mice.
Collapse
Affiliation(s)
- Yunqian Guan
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China; Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Role of human herpesvirus 8 interleukin-6-activated gp130 signal transducer in primary effusion lymphoma cell growth and viability. J Virol 2013; 87:10816-27. [PMID: 23903842 DOI: 10.1128/jvi.02047-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human herpesvirus 8 (HHV-8) infection is associated with Kaposi's sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman's disease. HHV-8-encoded viral interleukin-6 (vIL-6) is believed to contribute to pathogenesis via proproliferative, antiapoptotic, and proangiogenic activities. In PEL cells, vIL-6 is produced in functional amounts during viral latency and promotes the growth of these cells, mediating its activity from the endoplasmic reticulum (ER), where it is predominantly localized. This vIL-6 activity is dependent, in part, on its interaction with a splice variant of vitamin K epoxide reductase complex subunit 1 (VKORC1), termed VKORC1 variant 2 (VKORC1v2). Here we report that the IL-6 signal transducer, gp130, which can support vIL-6 signaling from the ER, is also required for optimal PEL cell growth and viability. Levels of activated extracellular regulated kinases (ERKs) 1 and 2 and signal transducer and activator of transcription 1 (STAT1) and STAT3, phosphorylated following gp130 stimulation, were reduced in gp130-depleted BCBL-1 and BC-1 cells. Diminished STAT activation was also detected in JSC-1 and BC-3 cells. Effects of gp130 depletion on growth could be mimicked by short hairpin RNA targeting of ERKs 1 and 2 or by depletion of STAT3. Finally, inhibition of vIL-6-gp130 association specifically within the ER compartment suppressed cell proliferation and viability, mirroring the effects of gp130 depletion. Combined, these data demonstrate that gp130, in addition to VKORC1v2, is essential for normal PEL cell growth and survival and that ER-localized vIL-6-gp130 interactions are critical for these activities. Targeting of intracellular vIL-6-gp130 interactions could potentially provide a means of PEL therapy.
Collapse
|
20
|
Regulated expression of lentivirus-mediated GDNF in human bone marrow-derived mesenchymal stem cells and its neuroprotection on dopaminergic cells in vitro. PLoS One 2013; 8:e64389. [PMID: 23717608 PMCID: PMC3661514 DOI: 10.1371/journal.pone.0064389] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 04/12/2013] [Indexed: 12/13/2022] Open
Abstract
Gene regulation remains one of the major challenges for gene therapy in clinical trials. In the present study, we first generated a binary tetracycline-on (Tet-On) system based on two lentivirus vectors, one expressing both human glial cell line-derived neurotrophic factor (hGDNF) and humanized recombinant green fluorescent protein (hrGFP) genes under second-generation tetracycline response element (TRE), and the other expressing the advanced reverse tetracycline-controlled transactivator--rtTA2S-M2 under a human minimal cytomegalovirus immediate early (CMV-IE) promoter. This system allows simultaneous expression of hGDNF and hrGFP genes in the presence of doxycycline (Dox). Human bone marrow-derived mesenchymal stem cells (hMSCs) were transduced with the binary Tet-On lentivirus vectors and characterized in vitro in the presence (On) or absence (Off) of Dox. The expression of hGDNF and hrGFP transgenes in transduced hMSCs was tightly regulated as determined by flow cytometry (FCM), GDNF enzyme-linked immunosorbent assay (ELISA) and quantitative real time-polymerase chain reaction (qRT-PCR). There was a dose-dependent regulation for hrGFP transgene expression. The levels of hGDNF protein in culture medium were correlated with the mean fluorescence intensity (MFI) units of hrGFP. The levels of transgene background expression were very low in the absence of Dox. The treatment of the conditioned medium from cultures of transduced hMSCs in the presence of Dox protected SH-SY5Y cells against 6-hydroxydopamine (6-OHDA) toxicity as determined by cell viability using 3, [4,5-dimethylthiazol-2-yl]-diphenyltetrazolium bromide (MTT) assay. The treatment of the conditioned medium was also found to improve the survival of dopaminergic (DA) neurons of ventral mesencephalic (VM) tissue in serum-free culture conditions as assessed by cell body area, the number of neurites and dendrite branching points, and proportion of tyrosine hydroxylase (TH)-immunoreactive (IR) cells. Our inducible lentivirus-mediated hGDNF gene delivery system may provide useful tools for basic research on gene therapy for chronic neurological disorders such as Parkinson's disease (PD).
Collapse
|
21
|
Rao L, Tang W, Wei Y, Bao L, Chen J, Chen H, He L, Lu P, Ren J, Wu L, Luan Z, Cui C, Xiao L. Highly efficient derivation of skeletal myotubes from human embryonic stem cells. Stem Cell Rev Rep 2013; 8:1109-19. [PMID: 23104134 DOI: 10.1007/s12015-012-9413-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Human embryonic stem cells (hESCs) are a promising model for the research of embryonic development and regenerative medicine. Since the first hESC line was established, many researchers have shown that pluripotent hESCs can be directed into many types of functional adult cells in culture. However, most of the reported methods have induced differentiation through the alteration of growth factors in the culture medium. These methods are time consuming; moreover, it is difficult to obtain a pure population of the desired cells because of the low efficiency of induction. In this study, we used a lentiviral-based inducible gene-expression system in hESCs to control the ectopic expression of MyoD, which is an essential transcription factor in skeletal muscle development. The induction of MyoD can efficiently direct the pluripotent hESCs into mesoderm in 24 h. The cells then become proliferated myoblasts and finally form multinucleated myotubes in vitro. The whole procedure took about 10 days, with an induction efficiency of over 90%. To our knowledge, this is the first time that hESCs have been induced into terminally differentiated cells with only one factor. In the future, these results could be a potential resource for cell therapy for diseases of muscle dysfunction.
Collapse
Affiliation(s)
- Lingjun Rao
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ellerström C, Strehl R, Hyllner J. Labeled stem cells as disease models and in drug discovery. Methods Mol Biol 2013; 997:239-51. [PMID: 23546761 DOI: 10.1007/978-1-62703-348-0_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Human pluripotent stem cells provide unique possibilities for in vitro studies of human cells in basic research, disease modeling as well as in industrial applications. By introducing relevant genome engineering technology, and thereby creating, for example, reporter cell lines, one will facilitate and improve safety pharmacology, toxicity testing, and can help the scientists to better understand pathological processes in humans. This review discusses how the merger of these two fields, human pluripotent stem cells and genome engineering, form extremely powerful tools and how they have been implemented already within the scientific community. In sharp contrast to immortalized human cell lines, which are both easy to expand and very simple to transfect, the genetically modified pluripotent stem cell line can be directed to a specific cell lineage and provide the user with highly relevant information. We highlight some of the challenges the field had to solve and how new technology advancements has removed the early bottlenecks.
Collapse
|
23
|
Inducible regulation of GDNF expression in human neural stem cells. SCIENCE CHINA-LIFE SCIENCES 2012; 56:32-9. [PMID: 23269553 DOI: 10.1007/s11427-012-4424-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/02/2012] [Indexed: 10/27/2022]
|
24
|
Kozlova EN, Berens C. Guiding Differentiation of Stem Cells in Vivo by Tetracycline-Controlled Expression of Key Transcription Factors. Cell Transplant 2012; 21:2537-54. [DOI: 10.3727/096368911x637407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transplantation of stem or progenitor cells is an attractive strategy for cell replacement therapy. However, poor long-term survival and insufficiently reproducible differentiation to functionally appropriate cells in vivo still present major obstacles for translation of this methodology to clinical applications. Numerous experimental studies have revealed that the expression of just a few transcription factors can be sufficient to drive stem cell differentiation toward a specific cell type, to transdifferentiate cells from one fate to another, or to dedifferentiate mature cells to pluripotent stem/progenitor cells (iPSCs). We thus propose here to apply the strategy of expressing the relevant key transcription factors to guide the differentiation of transplanted cells to the desired cell fate in vivo. To achieve this requires tools allowing us to control the expression of these genes in the transplant. Here, we describe drug-inducible systems that allow us to sequentially and timely activate gene expression from the outside, with a particular emphasis on the Tet system, which has been widely and successfully used in stem cells. These regulatory systems offer a tool for strictly limiting gene expression to the respective optimal stage after transplantation. This approach will direct the differentiation of the immature stem/progenitor cells in vivo to the desired cell type.
Collapse
Affiliation(s)
- Elena N Kozlova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
25
|
Abstract
This review aims to provide a broad overview of the targets, challenges and potential for gene therapy in the CNS, citing specific examples. There are a broad range of therapeutic targets, with very different requirements for a suitable viral vector. By utilizing different vector tropisms, novel routes of administration and engineered promoter control, transgenes can be targeted to specific therapeutic applications. Viral vectors have proven efficacious in preclinical models for several disease applications, spurring several clinical trials. While the field has pushed the limits of existing adeno-associated virus-based vectors, a next generation of vectors based on rational engineering of viral capsids should expand the application of gene therapy to be more effective in specific therapeutic applications.
Collapse
|
26
|
Laser-Based Propagation of Human iPS and ES Cells Generates Reproducible Cultures with Enhanced Differentiation Potential. Stem Cells Int 2012; 2012:926463. [PMID: 22701128 PMCID: PMC3369526 DOI: 10.1155/2012/926463] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 02/03/2012] [Accepted: 02/03/2012] [Indexed: 12/18/2022] Open
Abstract
Proper maintenance of stem cells is essential for successful utilization of ESCs/iPSCs as tools in developmental and drug discovery studies and in regenerative medicine. Standardization is critical for all future applications of stem cells and necessary to fully understand their potential. This study reports a novel approach for the efficient, consistent expansion of human ESCs and iPSCs using laser sectioning, instead of mechanical devices or enzymes, to divide cultures into defined size clumps for propagation. Laser-mediated propagation maintained the pluripotency, quality, and genetic stability of ESCs/iPSCs and led to enhanced differentiation potential. This approach removes the variability associated with ESC/iPSC propagation, significantly reduces the expertise, labor, and time associated with manual passaging techniques and provides the basis for scalable delivery of standardized ESC/iPSC lines. Adoption of standardized protocols would allow researchers to understand the role of genetics, environment, and/or procedural effects on stem cells and would ensure reproducible production of stem cell cultures for use in clinical/therapeutic applications.
Collapse
|
27
|
Avery S. Generation of inducible shRNAi human embryonic stem cell lines. ACTA ACUST UNITED AC 2012; Chapter 5:Unit5C.1. [PMID: 21913171 DOI: 10.1002/9780470151808.sc05c01s18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This unit describes the generation of tetracycline-inducible short hairpin RNA interference (shRNAi) human embryonic stem cell (hESC) lines. Using this vector-based approach enables stable and long-term expression of target hairpins under the control of doxycycline/tetracycline. Target degradation can be controlled in both a dose- and time-dependent manner that can even be switched off, depending upon the particular requirements of the study.
Collapse
|
28
|
Spontaneous transformation of adult mesenchymal stem cells from cynomolgus macaques in vitro. Exp Cell Res 2011; 317:2950-7. [PMID: 21963525 DOI: 10.1016/j.yexcr.2011.09.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Revised: 08/31/2011] [Accepted: 09/16/2011] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have shown potential clinical utility in cell therapy and tissue engineering, due to their ability to proliferate as well as to differentiate into multiple lineages, including osteogenic, adipogenic, and chondrogenic specifications. Therefore, it is crucial to assess the safety of MSCs while extensive expansion ex vivo is a prerequisite to obtain the cell numbers for cell transplantation. Here we show that MSCs derived from adult cynomolgus monkey can undergo spontaneous transformation following in vitro culture. In comparison with MSCs, the spontaneously transformed mesenchymal cells (TMCs) display significantly different growth pattern and morphology, reminiscent of the characteristics of tumor cells. Importantly, TMCs are highly tumorigenic, causing subcutaneous tumors when injected into NOD/SCID mice. Moreover, no multiple differentiation potential of TMCs is observed in vitro or in vivo, suggesting that spontaneously transformed adult stem cells may not necessarily turn into cancer stem cells. These data indicate a direct transformation of cynomolgus monkey MSCs into tumor cells following long-term expansion in vitro. The spontaneous transformation of the cultured cynomolgus monkey MSCs may have important implications for ongoing clinical trials and for models of oncogenesis, thus warranting a more strict assessment of MSCs prior to cell therapy.
Collapse
|
29
|
Chen CM, Krohn J, Bhattacharya S, Davies B. A comparison of exogenous promoter activity at the ROSA26 locus using a ΦiC31 integrase mediated cassette exchange approach in mouse ES cells. PLoS One 2011; 6:e23376. [PMID: 21853122 PMCID: PMC3154917 DOI: 10.1371/journal.pone.0023376] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/14/2011] [Indexed: 11/18/2022] Open
Abstract
The activities of nine ubiquitous promoters (ROSA26, CAG, CMV, CMVd1, UbC, EF1α, PGK, chicken β-actin and MC1) have been quantified and compared in mouse embryonic stem cells. To avoid the high variation in transgene expression which results from uncontrolled copy number and chromosomal position effects when using random insertion based transgenic approaches, we have adopted a PhiC31 integrase mediated cassette exchange method for the efficient insertion of transgenes at single copy within a defined and well characterized chromosomal position, ROSA26. This has enabled the direct comparison of constructs from within the same genomic context and allows a systematic and quantitative assessment of the strengths of the promoters in comparison with the endogenous ROSA26 promoter. The behavior of these exogenous promoters, when integrated at ROSA26 in both sense and antisense orientations, reveals a large variation in their levels of activity. In addition, a subset of promoters, EF1α, UbC and CAG, show an increased activity in the sense orientation as a consequence of integration. Transient transfection experiments confirmed these observations to reflect integration dependent effects and also revealed significant differences in the behaviour of these promoters when delivered transiently or stably. As well as providing an important reference which will facilitate the choice of an appropriate promoter to achieve the desired level of expression for a specific research question, this study also demonstrates the suitability of the cassette exchange methodology for the robust and reliable expression of multiple variant transgenes in ES cells.
Collapse
Affiliation(s)
- Chiann-mun Chen
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Jon Krohn
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Shoumo Bhattacharya
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Benjamin Davies
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
30
|
Affiliation(s)
- Yoon-Young Jang
- From the Sidney Kimmel Comprehensive Cancer Center; Stem Cell Program, Institute for Cell Engineering; and Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zhaohui Ye
- From the Sidney Kimmel Comprehensive Cancer Center; Stem Cell Program, Institute for Cell Engineering; and Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Linzhao Cheng
- From the Sidney Kimmel Comprehensive Cancer Center; Stem Cell Program, Institute for Cell Engineering; and Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
31
|
Han J, Sidhu K. Embryonic stem cell extracts: use in differentiation and reprogramming. Regen Med 2011; 6:215-27. [DOI: 10.2217/rme.11.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Stem cells have been studied extensively for decades and they have the inherent capacity to self-renew as well as to generate one or more types of specialized cells. The current focus of research on stem cells, particularly on embryonic stem cells, is on directed differentiation of these cells into specific cell types for future regenerative medicine. For the past few years, the process of reprogramming, which mediates convertion of somatic cells to their pluripotent state, has been given much attention, as it provides a possible source of autologous stem cells. In addition, understanding the molecular mechanism of differentiation and reprogramming has long been a subject of interest. In this article, we have briefly introduced stem cells and discussed the use of embryonic stem cells in reprogramming of somatic cells and differentiation to different lineages. The application of embryonic stem cells extracts in inducing reprogramming and transdifferentiation has also been described and discussed. Should this approach be successful, patient-specific cells will be produced safely and the likelihood of rejection will be decreased when used in cell therapy for many debilitating human diseases for which there is no cure such as Parkinson’s disease, Alzheimer’s disease, diabetes and others.
Collapse
Affiliation(s)
- Jinnuo Han
- Stem Cell Laboratory, School of Psychiatry, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Kuldip Sidhu
- Faculty of Medicine, Wallace Wurth Building, University of New South Wales, NSW 2052, Australia
| |
Collapse
|
32
|
Zhong Y, Sullenbarger B, Lasky LC. Effect of increased HoxB4 on human megakaryocytic development. Biochem Biophys Res Commun 2010; 398:377-82. [PMID: 20599537 PMCID: PMC2921174 DOI: 10.1016/j.bbrc.2010.06.075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 06/16/2010] [Indexed: 01/19/2023]
Abstract
In order to produce clinically useful quantities of platelets ex vivo we may need to firstly enhance early self-renewal of hematopoietic stem cells (HSCs) and/or megakaryocyte (Mk) progenitors. The homeodomain transcription factor HoxB4 has been shown to be an important regulator of stem cell renewal and hematopoiesis; however, its effect on megakaryopoiesis is unclear. In this study, we investigated the effect of HoxB4 overexpression or RNA silencing on megakaryocytic development in the human TF1 progenitor cell line; we then used recombinant tPTD-HoxB4 fusion protein to study the effect of exogenous HoxB4 on megakaryocytic development of human CD34 positively-selected cord blood cells. We found that ectopic HoxB4 in TF1 cells increased the antigen expression of CD61and CD41a, increased the gene expression of thrombopoietin receptor (TpoR), Scl-1, Cyclin D1, Fog-1 and Fli-1 while it decreased c-Myb expression. HoxB4 RNA silencing in TF1 cells decreased the expression of CD61 and CD41a and decreased Fli-1 expression while it increased the expression of c-Myb. Recombinant tPTD-HoxB4 fusion protein increased the percentages and absolute numbers of CD41a and CD61 positive cells during megakaryocytic differentiation of CD34 positively-selected cord blood cells and increased the numbers of colony-forming unit-megakaryocyte (CFU-Mk). Adding tPTD-HoxB4 fusion protein increased the gene expression of TpoR, Cyclin D1, Fog-1 and Fli-1 while it inhibited c-Myb expression. Our data suggest that increased HoxB4 enhanced early megakaryocytic development in human TF1 cells and CD34 positively-selected cord blood cells primarily by upregulating TpoR and Fli-1 expression and downregulating c-Myb expression. Increasing HoxB4 expression or adding recombinant HoxB4 protein might be a way to expand Mks for the production of platelets for use in transfusion medicine.
Collapse
Affiliation(s)
- Yiming Zhong
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
- Program in Molecular, Cellular, and Developmental Biology, The Ohio State University, Columbus, Ohio, USA
| | | | - Larry C. Lasky
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
- Program in Molecular, Cellular, and Developmental Biology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
33
|
Patsch C, Peitz M, Otte DM, Kesseler D, Jungverdorben J, Wunderlich FT, Brüstle O, Zimmer A, Edenhofer F. Engineering Cell-Permeant FLP Recombinase for Tightly Controlled Inducible and Reversible Overexpression in Embryonic Stem Cells. Stem Cells 2010; 28:894-902. [DOI: 10.1002/stem.417] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
34
|
A potent enhancer element in the 5′-UTR intron is crucial for transcriptional regulation of the human ubiquitin C gene. Gene 2009; 448:88-101. [DOI: 10.1016/j.gene.2009.08.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/26/2009] [Accepted: 08/28/2009] [Indexed: 02/01/2023]
|
35
|
Liu Y, Thyagarajan B, Lakshmipathy U, Xue H, Lieu P, Fontes A, MacArthur CC, Scheyhing K, Rao MS, Chesnut JD. Generation of Platform Human Embryonic Stem Cell Lines That Allow Efficient Targeting at a Predetermined Genomic Location. Stem Cells Dev 2009; 18:1459-72. [DOI: 10.1089/scd.2009.0047] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ying Liu
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Bhaskar Thyagarajan
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Uma Lakshmipathy
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Haipeng Xue
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Pauline Lieu
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Andrew Fontes
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Chad C. MacArthur
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Kelly Scheyhing
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Mahendra S. Rao
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Jonathan D. Chesnut
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| |
Collapse
|
36
|
Du ZW, Hu BY, Ayala M, Sauer B, Zhang SC. Cre recombination-mediated cassette exchange for building versatile transgenic human embryonic stem cells lines. Stem Cells 2009; 27:1032-41. [PMID: 19415769 PMCID: PMC2801346 DOI: 10.1002/stem.38] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
To circumvent the silencing effect of transgene expression in human embryonic stem cells (hESCs), we employed the Cre recombination-mediated cassette exchange strategy to target the silencing-resistant site in the genome. We have identified new loci that sustain transgene expression during stem cell expansion and differentiation to cells representing the three germ layers in vitro and in vivo. The built-in double loxP cassette in the established master hESC lines was specifically replaced by a targeting vector containing the same loxP sites, using the cell-permeable Cre protein transduction method, resulting in successful generation of new hESC lines with constitutive functional gene expression, inducible transgene expression, and lineage-specific reporter gene expression. This strategy and the master cell lines allow for rapid production of transgenic hESC lines in ordinary laboratories.
Collapse
Affiliation(s)
- Zhong-Wei Du
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, WiCell Institute, University of Wisconsin, Madison, WI, USA
| | - Bao-Yang Hu
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, WiCell Institute, University of Wisconsin, Madison, WI, USA
| | - Melvin Ayala
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, WiCell Institute, University of Wisconsin, Madison, WI, USA
| | - Brian Sauer
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Su-Chun Zhang
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, WiCell Institute, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
37
|
Pomper MG, Hammond H, Yu X, Ye Z, Foss CA, Lin DD, Fox JJ, Cheng L. Serial imaging of human embryonic stem-cell engraftment and teratoma formation in live mouse models. Cell Res 2009; 19:370-9. [PMID: 19114988 DOI: 10.1038/cr.2008.329] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Two new types of lentiviral vectors expressing a reporter transgene encoding either firefly luciferase (fLuc) for bioluminescence imaging or the HSV1 thymidine kinase (HSV1-TK) for radiopharmaceutical-based imaging were constructed to monitor human embryonic stem cell (hESC) engraftment and proliferation in live mice after transplantation. The constitutive expression of either transgene did not alter the properties of hESCs in the culture. We next monitored the formation of teratomas in SCID mice to test (1) whether the gene-modified hESCs maintain their developmental pluripotency, and (2) whether sustained reporter gene expression allows noninvasive, whole-body imaging of hESC derivatives in a live mouse model. We observed teratoma formation from both types of gene-modified cells as well as wild-type hESCs 2-4 months after inoculation. Using an optical imaging system, bioluminescence from the fLuc-transduced hESCs was easily detected in mice bearing teratomas long before palpable tumors could be detected. To develop a noninvasive imaging method more readily translatable to the clinic, we also utilized HSV1-TK and its specific substrate, 1-(2'-deoxy-2'-fluoro-beta-D-arabinofuranosyl)-5-[(125)I]iodouracil ([(125)I]FIAU), as a reporter/probe pair. After systemic administration, [(125)I]FIAU is phosphorylated only by the transgene-encoded HSV1-TK enzyme and retained within transduced (and transplanted) cells, allowing sensitive and quantitative imaging by single-photon emission computed tomography. Noninvasive imaging methods such as these may enable us to monitor the presence and distribution of transplanted human stem cells repetitively within live recipients over a long term through the expression of a reporter gene.
Collapse
Affiliation(s)
- Martin G Pomper
- Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Zafarana G, Avery SR, Avery K, Moore HD, Andrews PW. Specific knockdown of OCT4 in human embryonic stem cells by inducible short hairpin RNA interference. Stem Cells 2009; 27:776-82. [PMID: 19350677 PMCID: PMC2847189 DOI: 10.1002/stem.5] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 12/20/2008] [Indexed: 01/13/2023]
Abstract
Manipulation of gene function in embryonic stem cells by either over expression or downregulation is critical for understanding their subsequent cell fate. We have developed a tetracycline-inducible short hairpin RNA interference (shRNAi) for human embryonic stem cells (hESCs) and demonstrated doxycycline dose-dependent knockdown of the transcription factor OCT4 and the cell surface antigen beta2-microglobulin. The induced knockdown of OCT4 resulted in rapid differentiation of hESCs with a significant increase in transcription of genes associated with trophoblast and endoderm lineages, the extent of which was controlled by the degree of induction. Transgene toxicity, which may occur in conditional over-expression strategies with hESCs, was not observed with wild-type Tet repressor protein. The system allows efficient, reversible, and long-term downregulation of target genes in hESCs and enables the generation of stable transfectants for the knockdown of genes essential for cell survival and self-renewal, not necessarily possible by nonconditional shRNAi methods. STEM CELLS 2009;27:776-782.
Collapse
Affiliation(s)
- Gaetano Zafarana
- Department of Biomedical Science, Centre for Stem Cell Biology, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | | | | | | | | |
Collapse
|
39
|
Savage WJ, Barber JP, Mukhina GL, Hu R, Chen G, Matsui W, Thoburn C, Hess AD, Cheng L, Jones RJ, Brodsky RA. Glycosylphosphatidylinositol-anchored protein deficiency confers resistance to apoptosis in PNH. Exp Hematol 2009; 37:42-51. [PMID: 19013003 PMCID: PMC2628761 DOI: 10.1016/j.exphem.2008.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 08/28/2008] [Accepted: 09/02/2008] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Investigate the contribution of PIG-A mutations to clonal expansion in paroxysmal nocturnal hemoglobinuria (PNH). MATERIALS AND METHODS Primary CD34+ hematopoietic progenitors from PNH patients were assayed for annexin-V positivity by flow cytometry in a cell-mediated killing assay using autologous effectors from PNH patients or allogeneic effectors from healthy controls. To specifically assess the role of the PIG-A mutation in the development of clonal dominance and address confounders of secondary mutation and differential immune attack that can confound experiments using primary cells, we established an inducible PIG-A CD34+ myeloid cell line, TF-1. Apoptosis resistance was assessed after exposure to allogeneic effectors, NK92 cells (an interleukin-2-dependent cell line with the phenotype and function of activated natural killer [NK] cells), tumor necrosis factor (TNF)-alpha, and gamma-irradiation. Apoptosis was measured by annexin-V staining and caspase 3/7 activity. RESULTS In PNH patients, CD34+ hematopoietic progenitors lacking glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-AP(-)) were less susceptible than GPI-AP+ CD34+ precursors to autologous (8% vs 49%; p < 0.05) and allogeneic (28% vs 58%; p < 0.05) cell-mediated killing from the same patients. In the inducible PIG-A model, GPI-AP(-) TF-1 cells exhibited less apoptosis than induced, GPI-AP+ TF-1 cells in response to allogeneic cell-mediated killing, NK92-mediated killing, TNF-alpha, and gamma-irradiation. GPI-AP(-) TF-1 cells maintained resistance to apoptosis when effectors were raised against GPI-AP(-) cells, arguing against a GPI-AP being the target of immune attack in PNH. NK92-mediated killing was partially inhibited with blockade by specific antibodies to the stress-inducible GPI-AP ULBP1 and ULBP2 that activate immune effectors. Clonal competition experiments demonstrate that the mutant clone expands over time under proapoptotic conditions with TNF-alpha. CONCLUSION PIG-A mutations contribute to clonal expansion in PNH by conferring a survival advantage to hematopoietic progenitors under proapoptotic stresses.
Collapse
Affiliation(s)
- William J Savage
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Suter DM, Krause KH. Neural commitment of embryonic stem cells: molecules, pathways and potential for cell therapy. J Pathol 2008; 215:355-68. [PMID: 18566959 DOI: 10.1002/path.2380] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The study of neuronal differentiation of embryonic stem cells has raised major interest over recent years. It allows a better understanding of fundamental aspects of neurogenesis and, at the same time, the generation of neurons as tools for various applications ranging from drug testing to cell therapy and regenerative medicine. Since the first report of human embryonic stem (ES) cells derivation, many studies have shown the possibility of directing their differentiation towards neurons. However, there are still many challenges ahead, including gaining a better understanding of the mechanisms involved and developing techniques to allow the generation of homogeneous neuronal and glial subtypes. We review the current state of knowledge of embryonic neurogenesis which has been acquired from animal models and discuss its translation into in vitro strategies of neuronal differentiation of ES cells. We also highlight several aspects of current protocols which need to be optimized to generate high-quality embryonic stem cell-derived neuronal precursors suitable for clinical applications. Finally, we discuss the potential of embryonic stem cell-derived neurons for cell replacement therapy in several central nervous system diseases.
Collapse
Affiliation(s)
- D M Suter
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | |
Collapse
|
41
|
Mali P, Ye Z, Hommond HH, Yu X, Lin J, Chen G, Zou J, Cheng L. Improved efficiency and pace of generating induced pluripotent stem cells from human adult and fetal fibroblasts. Stem Cells 2008; 26:1998-2005. [PMID: 18511599 DOI: 10.1634/stemcells.2008-0346] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It was reported recently that human fibroblasts can be reprogrammed into a pluripotent state that resembles that of human embryonic stem (hES) cells. This was achieved by ectopic expression of four genes followed by culture on mouse embryonic fibroblast (MEF) feeders under a condition favoring hES cell growth. However, the efficiency of generating human induced pluripotent stem (iPS) cells is low, especially for postnatal human fibroblasts. We started supplementing with an additional gene or bioactive molecules to increase the efficiency of generating iPS cells from human adult as well as fetal fibroblasts. We report here that adding SV40 large T antigen (T) to either set of the four reprogramming genes previously used enhanced the efficiency by 23-70-fold from both human adult and fetal fibroblasts. Discernible hES-like colonies also emerged 1-2 weeks earlier if T was added. With the improved efficiency, we succeeded in replacing MEFs with immortalized human feeder cells that we previously established for optimal hES cell growth. We further characterized individually picked hES-like colonies after expansion (up to 24 passages). The majority of them expressed various undifferentiated hES markers. Some but not all the hES-like clones can be induced to differentiate into the derivatives of the three embryonic germ layers in both teratoma formation and embryoid body (EB) formation assays. These pluripotent clones also differentiated into trophoblasts after EB formation or bone morphogenetic protein 4 induction as classic hES cells. Using this improved approach, we also generated hES-like cells from homozygous fibroblasts containing the sickle cell anemia mutation Hemoglobin Sickle. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Prashant Mali
- Stem Cell Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Broadway Research Building, Room 747, 733 North Broadway, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Yu X, Zou J, Ye Z, Hammond H, Chen G, Tokunaga A, Mali P, Li YM, Civin C, Gaiano N, Cheng L. Notch signaling activation in human embryonic stem cells is required for embryonic, but not trophoblastic, lineage commitment. Cell Stem Cell 2008; 2:461-71. [PMID: 18462696 PMCID: PMC2442567 DOI: 10.1016/j.stem.2008.03.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 01/24/2008] [Accepted: 03/03/2008] [Indexed: 01/04/2023]
Abstract
The Notch signaling pathway plays important roles in cell-fate determination during embryonic development and adult life. In this study, we focus on the role of Notch signaling in governing cell-fate choices in human embryonic stem cells (hESCs). Using genetic and pharmacological approaches, we achieved both blockade and conditional activation of Notch signaling in several hESC lines. We report here that activation of Notch signaling is required for undifferentiated hESCs to form the progeny of all three embryonic germ layers, but not trophoblast cells. In addition, transient Notch signaling pathway activation enhanced generation of hematopoietic cells from committed hESCs. These new insights into the roles of Notch in hESC-fate determination may help to efficiently direct hESC differentiation into therapeutically relevant cell types.
Collapse
Affiliation(s)
- Xiaobing Yu
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jizhong Zou
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Zhaohui Ye
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Graduate Immunology Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Holly Hammond
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Guibin Chen
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Akinori Tokunaga
- Institute for Cell Engineering and Departments of Neurology, Neuroscience and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Prashant Mali
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Graduate Biomedical Engineering Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Yue-Ming Li
- Molecular Pharmacology & Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Curt Civin
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Nicholas Gaiano
- Institute for Cell Engineering and Departments of Neurology, Neuroscience and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Linzhao Cheng
- Stem Cell Program, Institute for Cell Engineering and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Graduate Immunology Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
43
|
Chen G, Ye Z, Yu X, Zou J, Mali P, Brodsky RA, Cheng L. Trophoblast differentiation defect in human embryonic stem cells lacking PIG-A and GPI-anchored cell-surface proteins. Cell Stem Cell 2008; 2:345-55. [PMID: 18397754 PMCID: PMC2442565 DOI: 10.1016/j.stem.2008.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 12/16/2007] [Accepted: 02/08/2008] [Indexed: 01/10/2023]
Abstract
Pluripotent human embryonic stem (hES) cells can differentiate into various cell types derived from the three embryonic germ layers and extraembryonic tissues such as trophoblasts. The mechanisms governing lineage choices of hES cells are largely unknown. Here, we report that we established two independent hES cell clones lacking a group of cell surface molecules, glycosyl-phosphatidyl-inositol-anchored proteins (GPI-APs). The GPI-AP deficiency in these two hES clones is due to the deficiency in the gene expression of PIG-A (phosphatidyl-inositol-glycan class A), which is required for the first step of GPI synthesis. GPI-AP-deficient hES cells were capable of forming embryoid bodies and initiating cell differentiation into the three embryonic germ layers. However, GPI-AP-deficient hES cells failed to form trophoblasts after differentiation induction by embryoid body formation or by adding exogenous BMP4. The defect in trophoblast formation was due to the lack of GPI-anchored BMP coreceptors, resulting in the impairment of full BMP4 signaling activation in the GPI-AP-deficient hES cells. These data reveal that GPI-AP-enhanced full activation of BMP signaling is required for human trophoblast formation.
Collapse
Affiliation(s)
- Guibin Chen
- Stem Cell Program, Institute for Cell Engineering, and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Zhaohui Ye
- Stem Cell Program, Institute for Cell Engineering, and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Graduate Program in Immunology, Johns Hopkins University, Baltimore, MD 21205
| | - Xiaobing Yu
- Stem Cell Program, Institute for Cell Engineering, and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jizhong Zou
- Stem Cell Program, Institute for Cell Engineering, and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Prashant Mali
- Stem Cell Program, Institute for Cell Engineering, and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Graduate Program in Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205
| | - Robert A. Brodsky
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Linzhao Cheng
- Stem Cell Program, Institute for Cell Engineering, and Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Graduate Program in Immunology, Johns Hopkins University, Baltimore, MD 21205
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
44
|
Fu JD, Jung Y, Chan CW, Li RA. An Inducible Transgene Expression System for Regulated Phenotypic Modification of Human Embryonic Stem Cells. Stem Cells Dev 2008; 17:315-24. [DOI: 10.1089/scd.2007.0114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ji-Dong Fu
- Stem Cell Program, University of California, Davis, CA 95616
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616
| | - Yunjoon Jung
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Camie W. Chan
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616
- Institute of Pediatric Regenerative Medicine, Shriners Hospital for Children of North America, Sacramento, CA 95817
| | - Ronald A. Li
- Stem Cell Program, University of California, Davis, CA 95616
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Institute of Pediatric Regenerative Medicine, Shriners Hospital for Children of North America, Sacramento, CA 95817
| |
Collapse
|
45
|
Xia X, Ayala M, Thiede BR, Zhang SC. In vitro- and in vivo-induced transgene expression in human embryonic stem cells and derivatives. Stem Cells 2008; 26:525-33. [PMID: 18032700 PMCID: PMC2707826 DOI: 10.1634/stemcells.2007-0710] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The use of human embryonic stem cells (hESCs) as a research and therapeutic tool will be facilitated by conditional gene expression. Here, we report drug-induced transgene expression, both in vitro and in vivo, from a tet-on hESC line with >95% purity. Using green fluorescent protein as an indicator, we demonstrated that the tet-on system allowed a tight control of the gene expression in both undifferentiated hESCs and differentiated cells of the three germ layers. More importantly, after the cells were transplanted into animals, the gene expression remained to be regulated by an orally administered drug. These results provide a technical basis for regulation of gene expression in hESCs and derivatives in vitro and in vivo.
Collapse
Affiliation(s)
- Xiaofeng Xia
- WiCell Research Institute, Madison, Wisconsin, USA
| | - Melvin Ayala
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Benjamin R. Thiede
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Su-Chun Zhang
- WiCell Research Institute, Madison, Wisconsin, USA
- Departments of Anatomy and Neurology, School of Medicine and Public Health, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
46
|
Lai Y, Drobinskaya I, Kolossov E, Chen C, Linn T. Genetic modification of cells for transplantation. Adv Drug Deliv Rev 2008; 60:146-59. [PMID: 18037530 DOI: 10.1016/j.addr.2007.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 08/02/2007] [Indexed: 01/16/2023]
Abstract
Progress in gene therapy has produced promising results that translate experimental research into clinical treatment. Gene modification has been extensively employed in cell transplantation. The main barrier is an effective gene delivery system. Several viral vectors were utilized in end-stage differentiated cells. Recently, successful applications were described with adenovirus-associated vectors. As an alternative, embryonic stem cell- and stem cell-like systems were established for generation of tissue-specified gene-modified cells. Owing to the feasibility for genetic manipulations and the self-renewing potency of these cells they can be used in a way enabling large-scale in vitro production. This approach offers the establishment of in vitro cell culture systems that will deliver sufficient amounts of highly purified, immunoautologous cells suitable for application in regenerative medicine. In this review, the current technology of gene delivery systems to cells is recapitulated and the latest developments for cell transplantation are discussed.
Collapse
|
47
|
Abstract
This chapter describes the methods we use to transduce mouse and human hematopoietic stem cells (HSCs) and human embryonic stem cells (hESCs). We provide detailed protocols for producing high-titer lentiviral supernatants by transient transfection and for measuring viral titers. Methods to concentrate viral supernatants to achieve a higher titer are also described. The protocols given here have been used successfully to transduce engrafting mouse and human HSCs as well as progenitor cells. These cells maintained stable transgene expression after engraftment in mice and in vivo differentiation. Human ESCs can also be transduced with a high efficiency, and transgene is expressed stably after hematopoietic differentiation.
Collapse
Affiliation(s)
- Zhaohui Ye
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | |
Collapse
|
48
|
Abstract
It has been demonstrated that several types of adult stem cells have a common attribute of tropism for gliomas. In our study, we provided evidence that embryonic stem cell-derived embryoid body (EB) cells also exhibited a tropism for gliomas. Chemotaxis assays and organotypic hippocampal slice culture experiments showed that EB cells were attracted by the conditioned medium from C6 glioma cells and by C6 glioma cells deposited on the slice. Aggregate culture assays showed that EB cells could coaggregate with C6 glioma cells. Embryoid body cells injected intratumorally were found to distribute throughout the tumor mass. All data indicated that EB cells displayed a tropism for gliomas.
Collapse
|
49
|
Thyagarajan B, Liu Y, Shin S, Lakshmipathy U, Scheyhing K, Xue H, Ellerström C, Strehl R, Hyllner J, Rao MS, Chesnut JD. Creation of engineered human embryonic stem cell lines using phiC31 integrase. Stem Cells 2007; 26:119-26. [PMID: 17962703 DOI: 10.1634/stemcells.2007-0283] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
It has previously been shown that the phage-derived phiC31 integrase can efficiently target native pseudo-attachment sites in the genome of various species in cultured cells, as well as in vivo. To demonstrate its utility in human embryonic stem cells (hESC), we have created hESC-derived clones containing expression constructs. Variant human embryonic stem cell lines BG01v and SA002 were used to derive lines expressing a green fluorescent protein (GFP) marker under control of either the human Oct4 promoter or the EF1alpha promoter. Stable clones were selected by antibiotic resistance and further characterized. The frequency of integration suggested candidate hot spots in the genome, which were mapped using a plasmid rescue strategy. The pseudo-attP profile in hESC differed from those reported earlier in differentiated cells. Clones derived using this method retained the ability to differentiate into all three germ layers, and fidelity of expression of GFP was verified in differentiation assays. GFP expression driven by the Oct4 promoter recapitulated endogenous Oct4 expression, whereas persistent stable expression of GFP expression driven by the EF1alpha promoter was seen. Our results demonstrate the utility of phiC31 integrase to target pseudo-attP sites in hESC and show that integrase-mediated site-specific integration can efficiently create stably expressing engineered human embryonic stem cell clones.
Collapse
|
50
|
Vieyra DS, Goodell MA. Pluripotentiality and Conditional Transgene Regulation in Human Embryonic Stem Cells Expressing Insulated Tetracycline-ON Transactivator. Stem Cells 2007; 25:2559-66. [PMID: 17628023 DOI: 10.1634/stemcells.2007-0248] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Conditional manipulation of gene expression by using tetracycline (TET)-ON based approaches has proven invaluable to study fundamental aspects of biology; however, the functionality of these systems in human embryonic stem cells (hESC) has not been established. Given the sensitivity of these cells to both genetic manipulation and variations of culture conditions, constitutive expression of TET transactivators might not only be toxic for hESC but might also impair their ability to self-renew or differentiate into multiple tissues. Therefore, the effect of these transactivators on the biology and pluripotentiality of hESC must first be evaluated before broad use of TET-ON methodologies is applied in these cells. Improved insulated lentivectors that display stable transgene expression and minimal insertional transactivation have been described for hESC. By using insulated lentivectors that allow simultaneous expression of TET components and fluorescent reporters, here we demonstrate that hESC constitutively expressing the TET-ON transactivator rtTA2SM2 can be derived and expanded in culture while retaining inducible transgene expression and pluripotentiality, including marker expression, a normal karyotype, and the ability to generate multiple tissues of different germ layer origin in teratomas. We also show that these cells retain the ability to control the expression of a stable integrated transgene in a doxycycline-dependent manner, which demonstrates that an insulated TET-ON lentiviral system is functional in hESC. Together, our results indicate that improved TET regulators like rtTA2SM2 in combination with insulated lentiviral-based systems offer alternative strategies for conditional gene expression in hESC. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Diego S Vieyra
- Stem Cells and Regenerative Medicine Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, N1030, Houston, Texas 77030, USA
| | | |
Collapse
|