1
|
Chu X, Hou Y, Peng C, Li W, Liang M, Mei J, Qian M, Wang J, Xu S, Jiang Y, Wen X, Chen Y, Yuan F, Xie J, Wang C, Zhang J. Exosome-derived miR-548ag drives hepatic lipid accumulation via upregulating FASN through inhibition of DNMT3B. J Lipid Res 2025:100818. [PMID: 40339699 DOI: 10.1016/j.jlr.2025.100818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of chronic liver disease worldwide. This study investigates the role of serum miR-548ag in regulating lipid metabolism and its contribution to MASLD in obesity. We found that miR-548ag levels were significantly elevated in the serum of both obese and MASLD patients, and positively correlated with body mass index (BMI), fasting plasma glucose (FPG), triglycerides (TG), total cholesterol (TC), LDL, HDL, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. Additionally, miR-548ag expression was significantly higher in the liver and abdominal adipose tissue of obese individuals compared to those of normal weight. In vitro studies in HepG2 and L02 cells, along with previous findings, demonstrated that miR-548ag promotes fatty acid synthase (FASN) expression by inhibiting DNA methyltransferase 3B (DNMT3B), thereby enhancing lipid synthesis. This was confirmed in two mouse models: one with tail vein injections of miR-548ag mimic/inhibitor adeno-associated viruses, and another with tail vein injections of exosomes from serum of normal-weight and obese individuals. Both models showed that miR-548ag upregulated FASN through DNMT3B inhibition, leading to increased lipid synthesis and larger hepatic lipid droplets, effects that were reversed by miR-548ag inhibition. Taken together, elevated miR-548ag expression in obesity enhances hepatic lipid synthesis by targeting DNMT3B to upregulate FASN, contributing to the development of MASLD.
Collapse
Affiliation(s)
- Xiaolong Chu
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Department of Medical Genetics , Medical College of Tarim University, 296 Tarim Avenue, Alar, Xinjiang, 843300, China
| | - Yanting Hou
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Chaoling Peng
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Wei Li
- First Affiliated Hospital of Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China
| | - Maodi Liang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Jin Mei
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Meiyu Qian
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Juan Wang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Shibo Xu
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Yidan Jiang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Xin Wen
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Yao Chen
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Fangyuan Yuan
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Jianxin Xie
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Department of Medical Genetics , Medical College of Tarim University, 296 Tarim Avenue, Alar, Xinjiang, 843300, China.
| | - Cuizhe Wang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China.
| | - Jun Zhang
- Medical College of Shihezi University, Bei-Er-Lu, Shihezi, Xinjiang, 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi, Xinjiang, 832000, China.
| |
Collapse
|
2
|
Sizer RE, Ingram RM, White RJ. Barriers Composed of tRNA Genes Can Complement the Benefits of a Ubiquitous Chromatin Opening Element to Enhance Transgene Expression. Biotechnol J 2025; 20:e202400455. [PMID: 39956936 PMCID: PMC11830863 DOI: 10.1002/biot.202400455] [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: 07/23/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 02/18/2025]
Abstract
Random integration of transgenes into host cell genomes often occurs in epigenetically unstable regions, leading to variable and unreliable transgene expression. To address this, biomanufacturing organizations frequently employ barrier elements, such as the widely-used ubiquitous chromatin opening element (UCOE). We have compared UCOE barrier activity against a barrier provided by tRNA genes. We demonstrate that the tRNA genes provide a more effective barrier than a UCOE in preventing transgene silencing in Chinese hamster ovary (CHO) cells. Nevertheless, the UCOE offers other benefits, increasing expression strongly, albeit transiently, and reducing production variability. Both the UCOE and tRNA genes counteract the repressive heterochromatin mark H3K9me3, but only the tRNA genes sustain euchromatic H3K27ac and recruitment of RNA polymerase II (Pol II) throughout long-term culture. A hybrid combining these distinct types of elements can provide benefits of both, enhancing expression in a more enduring manner. This synthetic hybrid offers potential for biomanufacturing applications.
Collapse
|
3
|
Zhang C, Liu D. Transcription Factor Binding Site in Promoter Determines the Pattern of Plasmid-Based Transgene Expression In Vivo. Pharmaceutics 2024; 16:544. [PMID: 38675205 PMCID: PMC11055139 DOI: 10.3390/pharmaceutics16040544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Understanding the regulation of transgene expression is critical for the success of plasmid-based gene therapy and vaccine development. In this study, we used two sets of plasmid vectors containing secreted embryonic alkaline phosphatase or the mouse IL-10 gene as a reporter and investigated the role of promoter elements in regulating transgene expression in vivo. We demonstrated in mice that hydrodynamic transfer of plasmids with the CMV promoter resulted in a high level of reporter gene expression that declined rapidly over time. In contrast, when plasmids with albumin promoters were used, a lower but sustained gene expression pattern was observed. We also found that plasmids containing a shorter CMV promoter sequence with fewer transcription factor binding sites showed a decrease in the peak level of gene expression without changing the overall pattern of reporter gene expression. The replacement of regulatory elements in the CMV promoter with a single regulatory element of the albumin promoter changed the pattern of transient gene expression seen in the CMV promoter to a pattern of sustained gene expression identical to that of a full albumin promoter. ChIP analyses demonstrated an elevated binding of acetylated histones and TATA box-binding protein to the promoter carrying regulatory elements of the albumin promoter. These results suggest that the strength of a promoter is determined by the number of appropriate transcription factor binding sites, while gene expression persistence is determined by the presence of regulatory elements capable of recruiting epigenetic modifying complexes that make the promoter accessible for transcription. This study provides important insights into the mechanisms underlying gene expression regulation in vivo, which can be used to improve plasmid-based gene therapy and vaccine development.
Collapse
Affiliation(s)
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602, USA;
| |
Collapse
|
4
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
5
|
Kim SM, Lee J, Lee JS. Implementation of ubiquitous chromatin opening elements as artificial integration sites for CRISPR/Cas9‐mediated knock‐in in mammalian cells. Eng Life Sci 2023; 23:e2200047. [PMID: 37025191 PMCID: PMC10071570 DOI: 10.1002/elsc.202200047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/16/2023] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
CRISPR/Cas9-mediated targeted gene integration (TI) has been used to generate recombinant mammalian cell lines with predictable transgene expression. Identifying genomic hot spots that render high and stable transgene expression and knock-in (KI) efficiency is critical for fully implementing TI-mediated cell line development (CLD); however, such identification is cumbersome. In this study, we developed an artificial KI construct that can be used as a hot spot at different genomic loci. The ubiquitous chromatin opening element (UCOE) was employed because of its ability to open chromatin and enable stable and site-independent transgene expression. UCOE KI cassettes were randomly integrated into CHO-K1 and HEK293T cells, followed by TI of enhanced green fluorescent protein (EGFP) onto the artificial UCOE KI site. The CHO-K1 random pool harboring 5'2.2A2UCOE-CMV displayed a significant increase in EGFP expression level and KI efficiency compared with that of the control without UCOE. In addition, 5'2.2A2UCOE-CMV showed improved Cas9 accessibility in the HEK293T genome, leading to an increase in indel frequency and homology-independent KI. Overall, this assessment revealed the potential of UCOE KI constructs as artificial integration sites in streamlining the screening of high-production targeted integrants by mitigating the selection of genomic hot spots.
Collapse
Affiliation(s)
- Seul Mi Kim
- Department of Molecular Science and Technology Ajou University Suwon Republic of Korea
| | - Jaejin Lee
- Department of Molecular Science and Technology Ajou University Suwon Republic of Korea
| | - Jae Seong Lee
- Department of Molecular Science and Technology Ajou University Suwon Republic of Korea
| |
Collapse
|
6
|
Gödecke N, Herrmann S, Weichelt V, Wirth D. A Ubiquitous Chromatin Opening Element and DNA Demethylation Facilitate Doxycycline-Controlled Expression during Differentiation and in Transgenic Mice. ACS Synth Biol 2023; 12:482-491. [PMID: 36755406 PMCID: PMC9942253 DOI: 10.1021/acssynbio.2c00450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Synthetic expression cassettes provide the ability to control transgene expression in experimental animal models through external triggers, enabling the study of gene function and the modulation of endogenous regulatory networks in vivo. The performance of synthetic expression cassettes in transgenic animals critically depends on the regulatory properties of the respective chromosomal integration sites, which are affected by the remodeling of the chromatin structure during development. The epigenetic status may affect the transcriptional activity of the synthetic cassettes and even lead to transcriptional silencing, depending on the chromosomal sites and the tissue. In this study, we investigated the influence of the ubiquitous chromosome opening element (UCOE) HNRPA2B1-CBX3 and its subfragments A2UCOE and CBX3 on doxycycline-controlled expression modules within the chromosomal Rosa26 locus. While HNRPA2B1-CBX3 and A2UCOE reduced the expression of the synthetic cassettes in mouse embryonic stem cells, CBX3 stabilized the expression and facilitated doxycycline-controlled expression after in vitro differentiation. In transgenic mice, the CBX3 element protected the cassettes from overt silencing although the expression was moderate and only partially controlled by doxycycline. We demonstrate that CBX3-flanked synthetic cassettes can be activated by decitabine-mediated blockade of DNA methylation or by specific recruitment of the catalytic demethylation domain of the ten-eleven translocation protein TET1 to the synthetic promoter. This suggests that CBX3 renders the synthetic cassettes permissive for subsequent epigenetic activation, thereby supporting doxycycline-controlled expression. Together, this study reveals a strategy for overcoming epigenetic constraints of synthetic expression cassettes, facilitating externally controlled transgene expression in mice.
Collapse
Affiliation(s)
- Natascha Gödecke
- RG
Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Sabrina Herrmann
- RG
Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Viola Weichelt
- RG
Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dagmar Wirth
- RG
Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany,Institute
of Experimental Hematology, Medical University
Hannover (MHH), 30625 Hannover, Germany,
| |
Collapse
|
7
|
Chu X, Hou Y, Zhang X, Li M, Ma D, Tang Y, Yuan C, Sun C, Liang M, Liu J, Wei Q, Chang Y, Wang C, Zhang J. Hepatic Glucose Metabolism Disorder Induced by Adipose Tissue-Derived miR-548ag via DPP4 Upregulation. Int J Mol Sci 2023; 24:ijms24032964. [PMID: 36769291 PMCID: PMC9917501 DOI: 10.3390/ijms24032964] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to explore the molecular mechanism underlying the regulation of glucose metabolism by miR-548ag. For the first time, we found that miR-548ag expression was elevated in the abdominal adipose tissue and serum of subjects with obesity and type 2 diabetes mellitus (T2DM). The conditional knockout of adipose tissue Dicer notably reduced the expression and content of miR-548ag in mouse adipose tissue, serum, and liver tissue. The combined use of RNAseq, an miRNA target gene prediction software, and the dual luciferase reporter assay confirmed that miR-548ag exerts a targeted regulatory effect on DNMT3B and DPP4. miR-548ag and DPP4 expression was increased in the adipose tissue, serum, and liver tissue of diet-induced obese mice, while DNMT3B expression was decreased. It was subsequently confirmed both in vitro and in vivo that adipose tissue-derived miR-548ag impaired glucose tolerance and insulin sensitivity by inhibiting DNMT3B and upregulating DPP4. Moreover, miR-548ag inhibitors significantly improved the adverse metabolic phenotype in both obese mice and db/db mice. These results revealed that the expression of the adipose tissue-derived miR-548ag increased in obese subjects, and that this could upregulate the expression of DPP4 by targeting DNMT3B, ultimately leading to glucose metabolism disorder. Therefore, miR-548ag could be utilized as a potential target in the treatment of T2DM.
Collapse
Affiliation(s)
- Xiaolong Chu
- Medical College, Shihezi University, Shihezi 832000, China
- Department of Medical Genetics, Medical College of Tarim University, Alaer 843300, China
| | - Yanting Hou
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Xueting Zhang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Menghuan Li
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Dingling Ma
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Yihan Tang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Chenggang Yuan
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Chaoyue Sun
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Maodi Liang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Jie Liu
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Qianqian Wei
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
| | - Yongsheng Chang
- Medical College, Shihezi University, Shihezi 832000, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Cuizhe Wang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
- Correspondence: (C.W.); (J.Z.); Tel./Fax: +86-993-205-5801 (C.W. & J.Z.)
| | - Jun Zhang
- Medical College, Shihezi University, Shihezi 832000, China
- Laboratory of Xinjiang Endemic and Ethic Diseases, Shihezi University, Shihezi 832000, China
- Correspondence: (C.W.); (J.Z.); Tel./Fax: +86-993-205-5801 (C.W. & J.Z.)
| |
Collapse
|
8
|
Cabrera A, Edelstein HI, Glykofrydis F, Love KS, Palacios S, Tycko J, Zhang M, Lensch S, Shields CE, Livingston M, Weiss R, Zhao H, Haynes KA, Morsut L, Chen YY, Khalil AS, Wong WW, Collins JJ, Rosser SJ, Polizzi K, Elowitz MB, Fussenegger M, Hilton IB, Leonard JN, Bintu L, Galloway KE, Deans TL. The sound of silence: Transgene silencing in mammalian cell engineering. Cell Syst 2022; 13:950-973. [PMID: 36549273 PMCID: PMC9880859 DOI: 10.1016/j.cels.2022.11.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/22/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
To elucidate principles operating in native biological systems and to develop novel biotechnologies, synthetic biology aims to build and integrate synthetic gene circuits within native transcriptional networks. The utility of synthetic gene circuits for cell engineering relies on the ability to control the expression of all constituent transgene components. Transgene silencing, defined as the loss of expression over time, persists as an obstacle for engineering primary cells and stem cells with transgenic cargos. In this review, we highlight the challenge that transgene silencing poses to the robust engineering of mammalian cells, outline potential molecular mechanisms of silencing, and present approaches for preventing transgene silencing. We conclude with a perspective identifying future research directions for improving the performance of synthetic gene circuits.
Collapse
Affiliation(s)
- Alan Cabrera
- Department of Bioengineering, Rice University, Houston, TX 77005, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hailey I Edelstein
- Center for Synthetic Biology, Northwestern University, Evanston, IL 60208, USA; The Eli and Edythe Broad CIRM Center, Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Fokion Glykofrydis
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033-9080, USA
| | - Kasey S Love
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sebastian Palacios
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Josh Tycko
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Meng Zhang
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, Urbana, IL 61801, USA
| | - Sarah Lensch
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Cara E Shields
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
| | - Mark Livingston
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Huimin Zhao
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, Urbana, IL 61801, USA
| | - Karmella A Haynes
- Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
| | - Leonardo Morsut
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033-9080, USA
| | - Yvonne Y Chen
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA 90095, USA
| | - Ahmad S Khalil
- Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Wilson W Wong
- Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - James J Collins
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033-9080, USA; Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Susan J Rosser
- School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Karen Polizzi
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK; Imperial College Centre for Synthetic Biology, South Kensington Campus, London, UK
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel 4058, Switzerland; Faculty of Science, University of Basel, Mattenstrasse 26, Basel 4058, Switzerland
| | - Isaac B Hilton
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Joshua N Leonard
- Center for Synthetic Biology, Northwestern University, Evanston, IL 60208, USA; The Eli and Edythe Broad CIRM Center, Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Lacramioara Bintu
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kate E Galloway
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tara L Deans
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
9
|
Use of ubiquitous chromatin opening elements (UCOE) as tools to maintain transgene expression in biotechnology. Comput Struct Biotechnol J 2022; 21:275-283. [PMID: 36582439 PMCID: PMC9764128 DOI: 10.1016/j.csbj.2022.11.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Amongst the most important outputs of the biopharmaceutical industry are recombinant proteins, many of which are produced by integrating transgenes into the genomes of mammalian cells. However, expression is highly variable and can be unstable during prolonged culture. This is often due to epigenetic mechanisms silencing the transgenes. To combat this problem, vectors have been engineered to include ubiquitous chromatin opening elements (UCOEs) that protect against silencing. Here, we recount the evidence that UCOEs can modify chromatin environments and benefit biomanufacturing.
Collapse
|
10
|
Episomes and Transposases-Utilities to Maintain Transgene Expression from Nonviral Vectors. Genes (Basel) 2022; 13:genes13101872. [PMID: 36292757 PMCID: PMC9601623 DOI: 10.3390/genes13101872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/04/2022] Open
Abstract
The efficient delivery and stable transgene expression are critical for applications in gene therapy. While carefully selected and engineered viral vectors allowed for remarkable clinical successes, they still bear significant safety risks. Thus, nonviral vectors are a sound alternative and avoid genotoxicity and adverse immunological reactions. Nonviral vector systems have been extensively studied and refined during the last decades. Emerging knowledge of the epigenetic regulation of replication and spatial chromatin organisation, as well as new technologies, such as Crispr/Cas, were employed to enhance the performance of different nonviral vector systems. Thus, nonviral vectors are in focus and hold some promising perspectives for future applications in gene therapy. This review addresses three prominent nonviral vector systems: the Sleeping Beauty transposase, S/MAR-based episomes, and viral plasmid replicon-based EBV vectors. Exemplarily, we review different utilities, modifications, and new concepts that were pursued to overcome limitations regarding stable transgene expression and mitotic stability. New insights into the nuclear localisation of nonviral vector molecules and the potential consequences thereof are highlighted. Finally, we discuss the remaining limitations and provide an outlook on possible future developments in nonviral vector technology.
Collapse
|
11
|
Wolff JH, Mikkelsen JG. Delivering genes with human immunodeficiency virus-derived vehicles: still state-of-the-art after 25 years. J Biomed Sci 2022; 29:79. [PMID: 36209077 PMCID: PMC9548131 DOI: 10.1186/s12929-022-00865-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 09/29/2022] [Indexed: 11/10/2022] Open
Abstract
Viruses are naturally endowed with the capacity to transfer genetic material between cells. Following early skepticism, engineered viruses have been used to transfer genetic information into thousands of patients, and genetic therapies are currently attracting large investments. Despite challenges and severe adverse effects along the way, optimized technologies and improved manufacturing processes are driving gene therapy toward clinical translation. Fueled by the outbreak of AIDS in the 1980s and the accompanying focus on human immunodeficiency virus (HIV), lentiviral vectors derived from HIV have grown to become one of the most successful and widely used vector technologies. In 2022, this vector technology has been around for more than 25 years. Here, we celebrate the anniversary by portraying the vector system and its intriguing properties. We dive into the technology itself and recapitulate the use of lentiviral vectors for ex vivo gene transfer to hematopoietic stem cells and for production of CAR T-cells. Furthermore, we describe the adaptation of lentiviral vectors for in vivo gene delivery and cover the important contribution of lentiviral vectors to basic molecular research including their role as carriers of CRISPR genome editing technologies. Last, we dwell on the emerging capacity of lentiviral particles to package and transfer foreign proteins.
Collapse
Affiliation(s)
- Jonas Holst Wolff
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark.
| |
Collapse
|
12
|
Seymour BJ, Singh S, Certo HM, Sommer K, Sather BD, Khim S, Clough C, Hale M, Pangallo J, Ryu BY, Khan IF, Adair JE, Rawlings DJ. Effective, safe, and sustained correction of murine XLA using a UCOE-BTK promoter-based lentiviral vector. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:635-651. [PMID: 33718514 PMCID: PMC7907679 DOI: 10.1016/j.omtm.2021.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
X-linked agammaglobulinemia (XLA) is an immune disorder caused by mutations in Bruton’s tyrosine kinase (BTK). BTK is expressed in B and myeloid cells, and its deficiency results in a lack of mature B cells and protective antibodies. We previously reported a lentivirus (LV) BTK replacement therapy that restored B cell development and function in Btk and Tec double knockout mice (a phenocopy of human XLA). In this study, with the goal of optimizing both the level and lineage specificity of BTK expression, we generated LV incorporating the proximal human BTK promoter. Hematopoietic stem cells from Btk−/−Tec−/− mice transduced with this vector rescued lineage-specific expression and restored B cell function in Btk−/−Tec−/− recipients. Next, we tested addition of candidate enhancers and/or ubiquitous chromatin opening elements (UCOEs), as well as codon optimization to improve BTK expression. An Eμ enhancer improved B cell rescue, but increased immunoglobulin G (IgG) autoantibodies. Addition of the UCOE avoided autoantibody generation while improving B cell development and function and reducing vector silencing. An optimized vector containing a truncated UCOE upstream of the BTK promoter and codon-optimized BTK cDNA resulted in stable, lineage-regulated BTK expression that mirrored endogenous BTK, making it a strong candidate for XLA therapy.
Collapse
Affiliation(s)
- Brenda J Seymour
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Swati Singh
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Hannah M Certo
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Blythe D Sather
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Socheath Khim
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Courtnee Clough
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Malika Hale
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Joseph Pangallo
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Byoung Y Ryu
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram F Khan
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jennifer E Adair
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medical Oncology, University of Washington, Seattle, WA 98195, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Departments of Pediatrics and Immunology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
13
|
Gödecke N, Herrmann S, Hauser H, Mayer-Bartschmid A, Trautwein M, Wirth D. Rational Design of Single Copy Expression Cassettes in Defined Chromosomal Sites Overcomes Intraclonal Cell-to-Cell Expression Heterogeneity and Ensures Robust Antibody Production. ACS Synth Biol 2021; 10:145-157. [PMID: 33382574 DOI: 10.1021/acssynbio.0c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The expression of endogenous genes as well as transgenes depends on regulatory elements within and surrounding genes as well as their epigenetic modifications. Members of a cloned cell population often show pronounced cell-to-cell heterogeneity with respect to the expression of a certain gene. To investigate the heterogeneity of recombinant protein expression we targeted cassettes into two preselected chromosomal hot-spots in Chinese hamster ovary (CHO) cells. Depending on the gene of interest and the design of the expression cassette, we found strong expression variability that could be reduced by epigenetic modifiers, but not by site-specific recruitment of the modulator dCas9-VPR. In particular, the implementation of ubiquitous chromatin opening elements (UCOEs) reduced cell-to-cell heterogeneity and concomitantly increased expression. The application of this method to recombinant antibody expression confirmed that rational design of cell lines for production of transgenes with predictable and high titers is a promising approach.
Collapse
Affiliation(s)
- Natascha Gödecke
- RG Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Sabrina Herrmann
- RG Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Hansjörg Hauser
- Staff Unit Scientific Strategy, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | | | | | - Dagmar Wirth
- RG Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
- Institute of Experimental Hematology, Medical University Hannover, Hannover 30625, Germany
| |
Collapse
|
14
|
Hoseinpoor R, Kazemi B, Rajabibazl M, Rahimpour A. Improving the expression of anti-IL-2Rα monoclonal antibody in the CHO cells through optimization of the expression vector and translation efficiency. J Biotechnol 2020; 324:112-120. [PMID: 33007349 DOI: 10.1016/j.jbiotec.2020.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
The growing need for monoclonal antibodies (mAbs) necessitates the development of novel and efficient production approaches. Regulatory elements like ubiquitous chromatin-opening elements (UCOEs) have been employed for improvement of the mAb expression in the Chinese hamster ovary (CHO) cells. SINEUPs are a class of long non-coding RNAs, which can improve the translation of partly overlapping mRNAs. A combination of these two elements might lead to higher production of mAbs. Therefore, the current study was conducted to investigate the effects of SINEUPs and A2UCOE on the expression of an IgG1 in the CHO-K1 cells. Hence, after constructing the mAb, mAb-SINEUP, and mAb-UCOE vectors, four stable cell pools were generated through combining the above vectors. According to the expression analysis, antibody yields were higher in the mAb-SINEUP and mAb-UCOE cell pools compared to the mAb cells. In addition, the cells possessing both SINEUP and UCOE elements provided the best expression. Persistent mAb expression was observed for over 2 months in these cells, whilst the expression was decreased in the mAb pool. SINEUP and UCOE positively influenced the stable mAb expression. It can be concluded that the SINEUP and UCOE enhance the antibody stability and expression level separately and their combination improves the mAb production in the CHO cells.
Collapse
Affiliation(s)
- Reyhaneh Hoseinpoor
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Ghosh R, Wood-Kaczmar A, Dobson L, Smith EJ, Sirinathsinghji EC, Kriston-Vizi J, Hargreaves IP, Heaton R, Herrmann F, Abramov AY, Lam AJ, Heales SJ, Ketteler R, Bates GP, Andre R, Tabrizi SJ. Expression of mutant exon 1 huntingtin fragments in human neural stem cells and neurons causes inclusion formation and mitochondrial dysfunction. FASEB J 2020; 34:8139-8154. [PMID: 32329133 PMCID: PMC8432155 DOI: 10.1096/fj.201902277rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 11/11/2022]
Abstract
Robust cellular models are key in determining pathological mechanisms that lead to neurotoxicity in Huntington's disease (HD) and for high throughput pre‐clinical screening of potential therapeutic compounds. Such models exist but mostly comprise non‐human or non‐neuronal cells that may not recapitulate the correct biochemical milieu involved in pathology. We have developed a new human neuronal cell model of HD, using neural stem cells (ReNcell VM NSCs) stably transduced to express exon 1 huntingtin (HTT) fragments with variable length polyglutamine (polyQ) tracts. Using a system with matched expression levels of exon 1 HTT fragments, we investigated the effect of increasing polyQ repeat length on HTT inclusion formation, location, neuronal survival, and mitochondrial function with a view to creating an in vitro screening platform for therapeutic screening. We found that expression of exon 1 HTT fragments with longer polyQ tracts led to the formation of intra‐nuclear inclusions in a polyQ length‐dependent manner during neurogenesis. There was no overt effect on neuronal viability, but defects of mitochondrial function were found in the pathogenic lines. Thus, we have a human neuronal cell model of HD that may recapitulate some of the earliest stages of HD pathogenesis, namely inclusion formation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Rhia Ghosh
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Alison Wood-Kaczmar
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Lucianne Dobson
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edward J Smith
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Eva C Sirinathsinghji
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Janos Kriston-Vizi
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | | | - Robert Heaton
- School of Pharmacy, Liverpool John Moores University, Liverpool, UK
| | | | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Amanda J Lam
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, London, UK
| | - Simon J Heales
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, London, UK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Gillian P Bates
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Ralph Andre
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
16
|
Schweickert PG, Cheng Z. Application of Genetic Engineering in Biotherapeutics Development. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09411-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
17
|
Gupta K, Parasnis M, Jain R, Dandekar P. Vector-related stratagems for enhanced monoclonal antibody production in mammalian cells. Biotechnol Adv 2019; 37:107415. [DOI: 10.1016/j.biotechadv.2019.107415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 07/01/2019] [Accepted: 07/01/2019] [Indexed: 12/16/2022]
|
18
|
Skipper KA, Hollensen AK, Antoniou MN, Mikkelsen JG. Sustained transgene expression from sleeping beauty DNA transposons containing a core fragment of the HNRPA2B1-CBX3 ubiquitous chromatin opening element (UCOE). BMC Biotechnol 2019; 19:75. [PMID: 31706316 PMCID: PMC6842454 DOI: 10.1186/s12896-019-0570-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022] Open
Abstract
Background DNA transposon-based vectors are effective nonviral tools for gene therapy and genetic engineering of cells. However, promoter DNA methylation and a near-random integration profile, which can result in transgene integration into heterochromatin, renders such vectors vulnerable to transcriptional repression. Therefore, to secure persistent transgene expression it may be necessary to protect transposon-embedded transgenes with anti-transcriptional silencing elements. Results We compare four different protective strategies in CHO-K1 cells. Our findings show robust protection from silencing of transgene cassettes mediated by the ubiquitous chromatin-opening element (UCOE) derived from the HNRPA2B1-CBX3 locus. Using a bioinformatic approach, we define a shorter HNRPA2B1-CBX3 UCOE core fragment and demonstrate that this can robustly maintain transgene expression after extended passaging of CHO-K1 cells carrying DNA transposon vectors equipped with this protective feature. Conclusions Our findings contribute to the understanding of the mechanism of HNRPA2B1-CBX3 UCOE-based transgene protection and support the use of a correctly oriented core fragment of this UCOE for DNA transposon vector-based production of recombinant proteins in CHO-K1 cells.
Collapse
Affiliation(s)
| | - Anne Kruse Hollensen
- Department of Biomedicine, HEALTH, Aarhus University, DK- 8000, Aarhus C, Denmark.,Department of Molecular Biology and Genetics, Science and Technology, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Michael N Antoniou
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, 8th Floor Tower Wing, Guy's Hospital, London, SE1 9RT, UK
| | | |
Collapse
|
19
|
Efficiency of different fragment lengths of the ubiquitous chromatin opening element HNRPA2B1-CBX3 in driving human CD18 gene expression within self-inactivating lentiviral vectors for gene therapy applications. Gene 2019; 710:265-272. [DOI: 10.1016/j.gene.2019.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 01/26/2023]
|
20
|
Zhu J, Hatton D. New Mammalian Expression Systems. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:9-50. [PMID: 28585079 DOI: 10.1007/10_2016_55] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are an increasing number of recombinant antibodies and proteins in preclinical and clinical development for therapeutic applications. Mammalian expression systems are key to enabling the production of these molecules, and Chinese hamster ovary (CHO) cell platforms continue to be central to delivery of the stable cell lines required for large-scale production. Increasing pressure on timelines and efficiency, further innovation of molecular formats and the shift to new production systems are driving developments of these CHO cell line platforms. The availability of genome and transcriptome data coupled with advancing gene editing tools are increasing the ability to design and engineer CHO cell lines to meet these challenges. This chapter aims to give an overview of the developments in CHO expression systems and some of the associated technologies over the past few years.
Collapse
Affiliation(s)
- Jie Zhu
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Diane Hatton
- MedImmune, Milstein Building, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
21
|
Zhang C, Zhang G, Liu D. Histone deacetylase inhibitors reactivate silenced transgene in vivo. Gene Ther 2018; 26:75-85. [DOI: 10.1038/s41434-018-0053-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/18/2018] [Accepted: 11/26/2018] [Indexed: 11/09/2022]
|
22
|
Cullmann K, Blokland KEC, Sebe A, Schenk F, Ivics Z, Heinz N, Modlich U. Sustained and regulated gene expression by Tet-inducible "all-in-one" retroviral vectors containing the HNRPA2B1-CBX3 UCOE ®. Biomaterials 2018; 192:486-499. [PMID: 30508767 DOI: 10.1016/j.biomaterials.2018.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 12/13/2022]
Abstract
Genetic modification of induced pluripotent stem (iPS) cells may be necessary for the generation of effector cells for cellular therapies. Hereby, it can be important to induce transgene expression at restricted and defined time windows, especially if it interferes with pluripotency or differentiation. To achieve this, inducible expression systems can be used such as the tetracycline-inducible retroviral vector system, however, retroviral expression can be subjected to epigenetic silencing or to position-effect variegation. One strategy to overcome this is the incorporation of ubiquitous chromatin opening elements (UCOE®'s) into retroviral vectors to maintain a transcriptionally permissive chromatin state at the integration site. In this study, we developed Tet-inducible all-in-one gammaretroviral vectors carrying different sized UCOE®'s derived from the A2UCOE. The ability to prevent vector silencing by preserving the Tet-regulatory potential was investigated in different cell lines, and in murine and human iPS cells. A 670-bp fragment spanning the CBX3 promoter region of A2UCOE (U670) was the most potent element in preventing silencing, and conferred the strongest expression from the vector in the induced state. While longer fragments of A2UCOEs also sustained expression, vector titers and induction efficiencies were impaired. Finally, we demonstrate that U670 can be used for constitutive expression of the transactivator in the all-in-one vector for faithful regulation of transgenes by doxycycline, including the thrombopoietin receptor Mpl conferring cytokine-dependent cell growth.
Collapse
Affiliation(s)
- Katharina Cullmann
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Kaj E C Blokland
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Attila Sebe
- Div. of Medical Biotechnology, Paul-Ehrlich-Institute, Langen, Germany
| | - Franziska Schenk
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany
| | - Zoltán Ivics
- Div. of Medical Biotechnology, Paul-Ehrlich-Institute, Langen, Germany
| | - Niels Heinz
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany; BioNTech Innovative Manufacturing Services GmbH, Idar-Oberstein, Germany
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, Div. of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Germany.
| |
Collapse
|
23
|
Cellular Antisilencing Elements Support Transgene Expression from Herpes Simplex Virus Vectors in the Absence of Immediate Early Gene Expression. J Virol 2018; 92:JVI.00536-18. [PMID: 29950408 DOI: 10.1128/jvi.00536-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/13/2018] [Indexed: 01/22/2023] Open
Abstract
Inactivation of all herpes simplex virus (HSV) immediate early (IE) genes to eliminate vector cytotoxicity results in rapid silencing of the viral genome, similar to the establishment of HSV latency. We recently reported that silencing of a nonviral reporter cassette could be overcome in nonneuronal cells by positioning the cassette in the viral latency (LAT) locus between resident chromatin boundary elements. Here, we tested the abilities of the chicken hypersensitive site 4 insulator and the human ubiquitous chromatin opening element A2UCOE to promote transgene expression from an IE-gene-inactivated HSV vector. We found that A2UCOE was particularly active in nonneuronal cells and reduced reporter promoter occupancy by a repressive histone mark. We determined whether multiple transgenes could be expressed under the control of different promoters from different loci of the same virus. The results showed abundant coexpression of LAT-embedded and A2UCOE-flanked genes in nonneuronal cells. In addition, a third reporter gene without known protective elements was active in cultured rat sensory neurons. These findings indicate that cellular antisilencing sequences can contribute to the expression of multiple genes from separate promoters in fully IE gene-disabled HSV vectors, providing an opportunity for therapeutic applications requiring mutually independent expression of different gene products from a single vector.IMPORTANCE Gene therapy has now entered a phase of development in which a growing number of recessive single gene defects can be successfully treated by vector-mediated introduction of a wild-type copy of the gene into the appropriate tissue. However, many disease conditions, such as neurodegeneration, cancer, and inflammatory processes, are more complex, requiring either multiple gene corrections or provision of coordinated gene activities to achieve a therapeutic outcome. Although herpes simplex virus (HSV) vectors have the capacity to meet this need, the challenge has been to genetically engineer the HSV genome in a manner to prevent expression of any viral genes while retaining the ability to express multiple therapeutic transgenes under independent transcriptional control. Here, we show that non-HSV insulator elements can be applied to retain at least transient transgene activity from multiple viral loci, thereby opening the door for more complex gene therapy applications in the future.
Collapse
|
24
|
Chaturvedi P, Zhao B, Zimmerman DL, Belmont AS. Stable and reproducible transgene expression independent of proliferative or differentiated state using BAC TG-EMBED. Gene Ther 2018; 25:376-391. [PMID: 29930343 PMCID: PMC6195848 DOI: 10.1038/s41434-018-0021-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/20/2018] [Accepted: 05/13/2018] [Indexed: 02/07/2023]
Abstract
Reproducible and stable transgene expression is an important goal in both basic research and biotechnology, with each application demanding a range of transgene expression. Problems in achieving stable transgene expression include multi-copy transgene silencing, chromosome-position effects, and loss of expression during long-term culture, induced cell quiescence, and/or cell differentiation. Previously, we described the “BAC TG-EMBED” method for copy-number dependent, chromosome position-independent expression of embedded transgenes within a BAC containing ~170 kb of the mouse Dhfr locus. Here we demonstrate wider applicability of the method by identifying a BAC and promoter combination that drives reproducible, copy-number dependent, position-independent transgene expression even after induced quiescence and/or cell differentiation into multiple cell types. Using a GAPDH BAC containing ~200 kb of the human GAPDH gene locus and a 1.2 kb human UBC promoter, we achieved stable GFP-ZeoR reporter expression in mouse NIH 3T3 cells after low-serum induced cell cycle arrest or differentiation into adipocytes. More notably, GFP-ZeoR expression remained stable and copy-number dependent even after differentiation of mouse ESCs into several distinct lineages. These results highlight the potential use of BAC TG-EMBED as an expression platform for high-level but stable, long-term expression of transgene independent of cell proliferative or differentiated state.
Collapse
Affiliation(s)
- Pankaj Chaturvedi
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL, USA
| | - Binhui Zhao
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL, USA
| | - David L Zimmerman
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL, USA.,Biology Department, College of the Ozarks, Point Lookout, MO, USA
| | - Andrew S Belmont
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
25
|
Romanova N, Noll T. Engineered and Natural Promoters and Chromatin-Modifying Elements for Recombinant Protein Expression in CHO Cells. Biotechnol J 2017; 13:e1700232. [DOI: 10.1002/biot.201700232] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/07/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Nadiya Romanova
- Cell Culture Technology; Faculty of Technology; Bielefeld University; Germany
| | - Thomas Noll
- Cell Culture Technology; Faculty of Technology; Bielefeld University; Germany
- Bielefeld University; Center for Biotechnology (CeBiTec); Germany
| |
Collapse
|
26
|
Kohlscheen S, Bonig H, Modlich U. Promises and Challenges in Hematopoietic Stem Cell Gene Therapy. Hum Gene Ther 2017; 28:782-799. [DOI: 10.1089/hum.2017.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Saskia Kohlscheen
- Research Group for Gene Modification in Stem Cells, Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, Langen, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Germany
- Department of Medicine/Division of Hematology, University of Washington, Seattle, Washington
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, Langen, Germany
| |
Collapse
|
27
|
Neville JJ, Orlando J, Mann K, McCloskey B, Antoniou MN. Ubiquitous Chromatin-opening Elements (UCOEs): Applications in biomanufacturing and gene therapy. Biotechnol Adv 2017; 35:557-564. [DOI: 10.1016/j.biotechadv.2017.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 12/31/2022]
|
28
|
Characterization of a core region in the A2UCOE that confers effective anti-silencing activity. Sci Rep 2017; 7:10213. [PMID: 28860464 PMCID: PMC5578987 DOI: 10.1038/s41598-017-10222-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/04/2017] [Indexed: 02/04/2023] Open
Abstract
We have previously shown that reliability of the A2UCOE in driving transgene expression can be attributed to its resistance to DNA methylation, and its ability to confer this property to linked regulatory sequences. In order to gain a better understanding of how resistance to DNA methylation from the A2UCOE is conferred, and whether the anti-silencing effect from the A2UCOE is confined within a core region, we evaluated the anti-silencing effect of different sub-domains. We found that maximal epigenetic regulatory activity was contained within a 455 bp element derived from the CBX3 region when tested in the context of a lentiviral vector in murine embryonic stem (ES) cells and human inducible pluripotent stem (iPS) cells. This region possessed an active chromatin signature, and operated effectively in cis to protect linked heterologous regulatory elements from methylation, thereby conferring stable transgene expression. Defined UCOE elements may be particularly useful for use in vectors where gene expression is desired in methylation-prone chromatin environments such as those encountered in pluripotent stem cells.
Collapse
|
29
|
The CpG-sites of the CBX3 ubiquitous chromatin opening element are critical structural determinants for the anti-silencing function. Sci Rep 2017; 7:7919. [PMID: 28801671 PMCID: PMC5554207 DOI: 10.1038/s41598-017-04212-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022] Open
Abstract
Suppression of therapeutic transgene expression from retroviral gene therapy vectors by epigenetic defence mechanisms represents a problem that is particularly encountered in pluripotent stem cells (PSCs) and their differentiated progeny. Transgene expression in these cells, however, can be stabilised by CpG-rich ubiquitous chromatin opening elements (UCOEs). In this context we recently demonstrated profound anti-silencing properties for the small (679 bp) CBX3-UCO element and we now confirmed this observation in the context of the defined murine chromosomal loci ROSA26 and TIGRE. Moreover, since the structural basis for the anti-silencing activity of UCOEs has remained poorly defined, we interrogated various CBX3 subfragments in the context of lentiviral vectors and murine PSCs. We demonstrated marked though distinct anti-silencing activity in the pluripotent state and during PSC-differentiation for several of the CBX3 subfragments. This activity was significantly correlated with CpG content as well as endogenous transcriptional activity. Interestingly, also a scrambled CBX3 version with preserved CpG-sites retained the anti-silencing activity despite the lack of endogenous promoter activity. Our data therefore highlight the importance of CpG-sites and transcriptional activity for UCOE functionality and suggest contributions from different mechanisms to the overall anti-silencing function of the CBX3 element.
Collapse
|
30
|
Ackermann M, Kuhn A, Kunkiel J, Merkert S, Martin U, Moritz T, Lachmann N. Ex vivo Generation of Genetically Modified Macrophages from Human Induced Pluripotent Stem Cells. Transfus Med Hemother 2017. [PMID: 28626364 DOI: 10.1159/000477129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Pluripotent stem cells, including induced pluripotent stem cells (iPSCs), have the capacity to differentiate towards all three germ layers and have been highlighted as an attractive cell source for the field of regenerative medicine. Thus, stable expression of therapeutic transgenes in iPSCs, as well as thereof derived progeny of hematopoietic lineage, may lay the foundation for innovative cell replacement therapies. METHODS We have utilized human iPSC lines genetically modified by lentiviral vector technology or targeted integration of reporter genes to evaluate transgene expression during hematopoietic specification and differentiation towards macrophages. RESULTS Use of lentiviral vectors equipped with an ubiquitous chromatin opening element (CBX3-UCOE) as well as zinc finger nuclease-mediated targeting of an expression cassette into the human adeno-associated virus integration site 1 (AAVS1) safe harbor resulted in stable transgene expression in iPSCs. When iPSCs were differentiated along the myeloid pathway into macrophages, both strategies yielded sustained transgene expression during the hematopoietic specification process including mature CD14+ and CD11b+ macrophages. CONCLUSION Combination of human iPSC technology with either lentiviral vector technology or designer nuclease-based genome editing allows for the generation of transgenic iPSC-derived macrophages with stable transgene expression which may be useful for novel cell and gene replacement therapies.
Collapse
Affiliation(s)
- Mania Ackermann
- JRG Translational Hematology, REBIRTH Cluster of Excellence, Hanover Medical School, Hanover, Germany.,Institute of Experimental Hematology, Hanover Medical School, Hanover, Germany
| | - Alexandra Kuhn
- Institute of Experimental Hematology, Hanover Medical School, Hanover, Germany.,RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hanover Medical School, Hanover, Germany
| | - Jessica Kunkiel
- Institute of Experimental Hematology, Hanover Medical School, Hanover, Germany.,RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hanover Medical School, Hanover, Germany
| | - Sylvia Merkert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Cluster of Excellence, Hanover Medical School, Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hanover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Cluster of Excellence, Hanover Medical School, Hanover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hanover, Germany
| | - Thomas Moritz
- Institute of Experimental Hematology, Hanover Medical School, Hanover, Germany.,RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hanover Medical School, Hanover, Germany
| | - Nico Lachmann
- JRG Translational Hematology, REBIRTH Cluster of Excellence, Hanover Medical School, Hanover, Germany.,Institute of Experimental Hematology, Hanover Medical School, Hanover, Germany
| |
Collapse
|
31
|
Chatzinikolaou G, Apostolou Z, Aid-Pavlidis T, Ioannidou A, Karakasilioti I, Papadopoulos GL, Aivaliotis M, Tsekrekou M, Strouboulis J, Kosteas T, Garinis GA. ERCC1-XPF cooperates with CTCF and cohesin to facilitate the developmental silencing of imprinted genes. Nat Cell Biol 2017; 19:421-432. [PMID: 28368372 DOI: 10.1038/ncb3499] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 02/24/2017] [Indexed: 12/15/2022]
Abstract
Inborn defects in DNA repair are associated with complex developmental disorders whose causal mechanisms are poorly understood. Using an in vivo biotinylation tagging approach in mice, we show that the nucleotide excision repair (NER) structure-specific endonuclease ERCC1-XPF complex interacts with the insulator binding protein CTCF, the cohesin subunits SMC1A and SMC3 and with MBD2; the factors co-localize with ATRX at the promoters and control regions (ICRs) of imprinted genes during postnatal hepatic development. Loss of Ercc1 or exposure to MMC triggers the localization of CTCF to heterochromatin, the dissociation of the CTCF-cohesin complex and ATRX from promoters and ICRs, altered histone marks and the aberrant developmental expression of imprinted genes without altering DNA methylation. We propose that ERCC1-XPF cooperates with CTCF and cohesin to facilitate the developmental silencing of imprinted genes and that persistent DNA damage triggers chromatin changes that affect gene expression programs associated with NER disorders.
Collapse
Affiliation(s)
- Georgia Chatzinikolaou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Zivkos Apostolou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| | - Tamara Aid-Pavlidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Anna Ioannidou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| | - Ismene Karakasilioti
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Giorgio L Papadopoulos
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
- Division of Molecular Oncology, Biomedical Sciences Research Center 'Alexander Fleming', GR 16672 Vari, Greece
| | - Michalis Aivaliotis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Maria Tsekrekou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| | - John Strouboulis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Division of Molecular Oncology, Biomedical Sciences Research Center 'Alexander Fleming', GR 16672 Vari, Greece
| | - Theodore Kosteas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Biology, University of Crete, Vassilika Vouton, GR71409 Heraklion, Crete, Greece
| |
Collapse
|
32
|
Detailed comparison of retroviral vectors and promoter configurations for stable and high transgene expression in human induced pluripotent stem cells. Gene Ther 2017; 24:298-307. [PMID: 28346436 DOI: 10.1038/gt.2017.20] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/27/2017] [Accepted: 03/06/2017] [Indexed: 12/19/2022]
Abstract
Correction of patient-specific induced pluripotent stem cells (iPSC) upon gene delivery through retroviral vectors offers new treatment perspectives for monogenetic diseases. Gene-modified iPSC clones can be screened for safe integration sites and differentiated into transplantable cells of interest. However, the current bottleneck is epigenetic vector silencing. In order to identify the most suitable retroviral expression system in iPSC, we systematically compared vectors from different retroviral genera, different promoters and their combination with ubiquitous chromatin opening elements (UCOE), and several envelope pseudotypes. Lentiviral vectors (LV) pseudotyped with vesicular stomatitis virus glycoprotein were superior to gammaretroviral and alpharetroviral vectors and other envelopes tested. The elongation factor 1α short (EFS) promoter mediated the most robust expression, whereas expression levels were lower from the potent but more silencing-prone spleen focus forming virus (SFFV) promoter. Both full-length (A2UCOE) and minimal (CBX3) UCOE juxtaposed to two physiological and one viral promoter reduced transgene silencing with equal efficiency. However, a promoter-specific decline in expression levels was not entirely prevented. Upon differentiation of transgene-positive iPSC into endothelial cells, A2UCOE.EFS and CBX3.EFS vectors maintained highest transgene expression in a larger fraction of cells as compared with all other constructs tested here. The function of UCOE diminished, but did not fully counteract, vector silencing and possibilities for improvements remain. Nevertheless, the CBX3.EFS in a LV background exhibited the most promising promoter and vector configuration for both high titer production and long-term genetic modification of human iPSC and their progeny.
Collapse
|
33
|
Nematpour F, Mahboudi F, Vaziri B, Khalaj V, Ahmadi S, Ahmadi M, Ebadat S, Davami F. Evaluating the expression profile and stability of different UCOE containing vector combinations in mAb-producing CHO cells. BMC Biotechnol 2017; 17:18. [PMID: 28228095 PMCID: PMC5322649 DOI: 10.1186/s12896-017-0330-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/03/2017] [Indexed: 12/03/2022] Open
Abstract
Background As the demand for monoclonal antibodies (mAb) increases, more efficient expression methods are required for their manufacturing process. Transcriptional gene silencing is a common phenomenon in recombinant cell lines which leads to expression reduction and instability. There are reports on improved antibody expression in ubiquitous chromatin opening element (UCOE) containing both heavy and light chain gene constructs. Here we investigate the impact of having these elements as part of the light chain, heavy chain or both genes during cell line development. In this regard, non-UCOE and UCOE vectors were constructed and stable Chinese hamster ovary (CHO) cell pools were generated by different vector combinations. Results Expression analysis revealed that all UCOE cell pools had higher antibody yields compared to non-UCOE cells, Moreover the most optimal expression was obtained by cells containing just the UCOE on heavy chain. In terms of stability, it was shown that the high level of expression was kept consistence for more than four months in these cells whereas the expression titers were reduced in the other UCOE pools. Conclusions In conclusion, UCOE significantly enhanced the level and stability of antibody expression and the use of this element with heavy chain provided more stable cell lines with higher production level.
Collapse
Affiliation(s)
- Fatemeh Nematpour
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fereidoun Mahboudi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Behrouz Vaziri
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Vahid Khalaj
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Samira Ahmadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Maryam Ahmadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.,Departments of Medical Biotechnology, Semnan University of Medical Sciences, Semnan, 3519899951, Iran
| | - Saedeh Ebadat
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
34
|
Maetzig T, Ruschmann J, Lai CK, Ngom M, Imren S, Rosten P, Norddahl GL, von Krosigk N, Sanchez Milde L, May C, Selich A, Rothe M, Dhillon I, Schambach A, Humphries RK. A Lentiviral Fluorescent Genetic Barcoding System for Flow Cytometry-Based Multiplex Tracking. Mol Ther 2017; 25:606-620. [PMID: 28253481 DOI: 10.1016/j.ymthe.2016.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
Retroviral integration site analysis and barcoding have been instrumental for multiplex clonal fate mapping, although their use imposes an inherent delay between sample acquisition and data analysis. Monitoring of multiple cell populations in real time would be advantageous, but multiplex assays compatible with flow cytometric tracking of competitive growth behavior are currently limited. We here describe the development and initial validation of three generations of lentiviral fluorescent genetic barcoding (FGB) systems that allow the creation of 26, 14, or 6 unique labels. Color-coded populations could be tracked in multiplex in vitro assays for up to 28 days by flow cytometry using all three vector systems. Those involving lower levels of multiplexing eased color-code generation and the reliability of vector expression and enabled functional in vitro and in vivo studies. In proof-of-principle experiments, FGB vectors facilitated in vitro multiplex screening of microRNA (miRNA)-induced growth advantages, as well as the in vivo recovery of color-coded progeny of murine and human hematopoietic stem cells. This novel series of FGB vectors provides new tools for assessing comparative growth properties in in vitro and in vivo multiplexing experiments, while simultaneously allowing for a reduction in sample numbers by up to 26-fold.
Collapse
Affiliation(s)
- Tobias Maetzig
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany.
| | - Jens Ruschmann
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Courteney K Lai
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Mor Ngom
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Suzan Imren
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Patricia Rosten
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Gudmundur L Norddahl
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Niklas von Krosigk
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Lea Sanchez Milde
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Christopher May
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Anton Selich
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Ishpreet Dhillon
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - R Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
35
|
NEMATPOUR F, MAHBOUDI F, KHALAJ V, VAZIRI B, AHMADI S, AHMADI M, EBADAT S, DAVAMI F. Optimization of monoclonal antibody expression in CHOcells by employing epigenetic gene regulation tools. Turk J Biol 2017. [DOI: 10.3906/biy-1702-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
36
|
Hoepfner J, Kleinsorge M, Papp O, Ackermann M, Alfken S, Rinas U, Solodenko W, Kirschning A, Sgodda M, Cantz T. Biphasic modulation of Wnt signaling supports efficient foregut endoderm formation from human pluripotent stem cells. Cell Biol Int 2016; 40:534-48. [DOI: 10.1002/cbin.10590] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/07/2016] [Indexed: 01/12/2023]
Affiliation(s)
- Jeannine Hoepfner
- Translational Hepatology and Stem Cell Biology; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
- Department of Gastroenterology, Hepatology, and Endocrinology; Hannover Medical School; Hannover Germany
| | - Mandy Kleinsorge
- Translational Hepatology and Stem Cell Biology; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
- Department of Gastroenterology, Hepatology, and Endocrinology; Hannover Medical School; Hannover Germany
| | - Oliver Papp
- Translational Hepatology and Stem Cell Biology; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
- Department of Gastroenterology, Hepatology, and Endocrinology; Hannover Medical School; Hannover Germany
| | - Mania Ackermann
- iPSC Based Gene Therapy; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
| | - Susanne Alfken
- Translational Hepatology and Stem Cell Biology; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
- Department of Gastroenterology, Hepatology, and Endocrinology; Hannover Medical School; Hannover Germany
| | - Ursula Rinas
- Institute of Technical Chemistry; Leibniz University Hannover; Hannover Germany
| | - Wladimir Solodenko
- Institute of Organic Chemistry; Leibniz University Hannover; Hannover Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry; Leibniz University Hannover; Hannover Germany
| | - Malte Sgodda
- Translational Hepatology and Stem Cell Biology; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
- Department of Gastroenterology, Hepatology, and Endocrinology; Hannover Medical School; Hannover Germany
| | - Tobias Cantz
- Translational Hepatology and Stem Cell Biology; REBIRTH Cluster of Excellence, Hannover Medical School; Hannover Germany
- Department of Gastroenterology, Hepatology, and Endocrinology; Hannover Medical School; Hannover Germany
- Cell and Developmental Biology; Max Planck Institute for Molecular Biomedicine; Münster Germany
| |
Collapse
|
37
|
Krinner S, Heitzer A, Asbach B, Wagner R. Interplay of Promoter Usage and Intragenic CpG Content: Impact on GFP Reporter Gene Expression. Hum Gene Ther 2015; 26:826-40. [PMID: 26414116 DOI: 10.1089/hum.2015.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Successful therapeutic protein production in vitro and in vivo requires efficient and long-term transgene expression supported by optimized vector and transgene cis-regulatory sequence elements. This study provides a comparative analysis of CpG-rich, highly expressed, versus CpG-depleted, poorly expressed green fluorescent protein (GFP) reporter transgenes, transcribed by various promoters in two different cell systems. Long-term GFP expression from a defined locus in stable Chinese hamster ovary cells was clearly influenced by the combination of transgene CpG content and promoter usage, as shown by differential silencing effects on selection pressure removal among the cytomegalovirus (CMV) promoter and elongation factor (EF)-1α promoter. Whereas a high intragenic CpG content promoted local DNA methylation, CpG depletion rather accelerated transgene loss and increased the local chromatin density. On lentiviral transfer of various expression modules into epigenetically sensitive P19 embryonic pluripotent carcinoma cells, CMV promoter usage led to rapid gene silencing irrespective of the intragenic CpG content. In contrast, EF-1α promoter-controlled constructs showed delayed silencing activity and high-level transgene expression, in particular when the CpG-rich GFP reporter was used. Notably, GFP silencing in P19 cells could be prevented completely by the bidirectional, dual divergently transcribed A2UCOE (ubiquitously acting chromatin-opening element derived from the human HNRPA2B1-CBX3 locus) promoter. Because the level of GFP expression by the A2UCOE promoter was entirely unaffected by the intragenic CpG level, we suggest that A2UCOE can overcome chromatin compaction resulting from intragenic CpG depletion due to its ascribed chromatin-opening abilities. Our analyses provide insights into the interplay of the intragenic CpG content with promoter sequences and regulatory sequence elements, thus contributing toward the design of therapeutic transgene expression cassettes for future gene therapy applications.
Collapse
Affiliation(s)
- Simone Krinner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology and Gene Therapy Unit, University of Regensburg , Regensburg, Germany
| | - Asli Heitzer
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology and Gene Therapy Unit, University of Regensburg , Regensburg, Germany
| | - Benedikt Asbach
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology and Gene Therapy Unit, University of Regensburg , Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology and Gene Therapy Unit, University of Regensburg , Regensburg, Germany
| |
Collapse
|
38
|
Harraghy N, Calabrese D, Fisch I, Girod PA, LeFourn V, Regamey A, Mermod N. Epigenetic regulatory elements: Recent advances in understanding their mode of action and use for recombinant protein production in mammalian cells. Biotechnol J 2015; 10:967-78. [DOI: 10.1002/biot.201400649] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/20/2015] [Accepted: 05/20/2015] [Indexed: 12/18/2022]
|
39
|
Betts Z, Dickson AJ. Assessment of UCOE on Recombinant EPO Production and Expression Stability in Amplified Chinese Hamster Ovary Cells. Mol Biotechnol 2015; 57:846-58. [DOI: 10.1007/s12033-015-9877-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
40
|
Zboray K, Sommeregger W, Bogner E, Gili A, Sterovsky T, Fauland K, Grabner B, Stiedl P, Moll HP, Bauer A, Kunert R, Casanova E. Heterologous protein production using euchromatin-containing expression vectors in mammalian cells. Nucleic Acids Res 2015; 43:e102. [PMID: 25977298 PMCID: PMC4652741 DOI: 10.1093/nar/gkv475] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/29/2015] [Indexed: 01/10/2023] Open
Abstract
Upon stable cell line generation, chromosomal integration site of the vector DNA has a major impact on transgene expression. Here we apply an active gene environment, rather than specified genetic elements, in expression vectors used for random integration. We generated a set of Bacterial Artificial Chromosome (BAC) vectors with different open chromatin regions, promoters and gene regulatory elements and tested their impact on recombinant protein expression in CHO cells. We identified the Rosa26 BAC as the most efficient vector backbone showing a nine-fold increase in both polyclonal and clonal production of the human IgG-Fc. Clonal protein production was directly proportional to integrated vector copy numbers and remained stable during 10 weeks without selection pressure. Finally, we demonstrated the advantages of BAC-based vectors by producing two additional proteins, HIV-1 glycoprotein CN54gp140 and HIV-1 neutralizing PG9 antibody, in bioreactors and shake flasks reaching a production yield of 1 g/l.
Collapse
Affiliation(s)
- Katalin Zboray
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, 1090, Austria
| | - Wolfgang Sommeregger
- Vienna Institute of BioTechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190, Austria Polymun Scientific GmbH, Klosterneuburg, 3400, Austria
| | - Edith Bogner
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, 1090, Austria
| | - Andreas Gili
- Polymun Scientific GmbH, Klosterneuburg, 3400, Austria
| | | | | | - Beatrice Grabner
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, 1090, Austria
| | - Patricia Stiedl
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, 1090, Austria
| | - Herwig P Moll
- Institute of Pharmacology, Center of Physiology and Pharmacology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
| | | | - Renate Kunert
- Vienna Institute of BioTechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Emilio Casanova
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, 1090, Austria Institute of Pharmacology, Center of Physiology and Pharmacology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, 1090, Austria
| |
Collapse
|
41
|
Betts Z, Croxford AS, Dickson AJ. Evaluating the interaction between UCOE and DHFR-linked amplification and stability of recombinant protein expression. Biotechnol Prog 2015; 31:1014-25. [DOI: 10.1002/btpr.2083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/17/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Zeynep Betts
- Faculty of Life Sciences; University of Manchester; Michael Smith Building, Oxford Road Manchester M13 9PT UK
| | - Alexandra S Croxford
- Faculty of Life Sciences; University of Manchester; Michael Smith Building, Oxford Road Manchester M13 9PT UK
| | - Alan J Dickson
- Faculty of Life Sciences; University of Manchester; Michael Smith Building, Oxford Road Manchester M13 9PT UK
| |
Collapse
|
42
|
Chromatin function modifying elements in an industrial antibody production platform--comparison of UCOE, MAR, STAR and cHS4 elements. PLoS One 2015; 10:e0120096. [PMID: 25849659 PMCID: PMC4388700 DOI: 10.1371/journal.pone.0120096] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/02/2015] [Indexed: 01/02/2023] Open
Abstract
The isolation of stably transfected cell lines suitable for the manufacture of biotherapeutic protein products can be an arduous process relying on the identification of a high expressing clone; this frequently involves transgene amplification and maintenance of the clones' expression over at least 60 generations. Maintenance of expression, or cell line stability, is highly dependent upon the nature of the genomic environment at the site of transgene integration, where epigenetic mechanisms lead to variable expression and silencing in the vast majority of cases. We have assessed four chromatin function modifying elements (A2UCOE, MAR X_S29, STAR40 and cHS4) for their ability to negate chromatin insertion site position effects and their ability to express and maintain monoclonal antibody expression. Each element was analysed by insertion into different positions within a vector, either flanking or between heavy chain (HC) and light chain (LC) antibody expression cassettes. Our results clearly show that the A2UCOE is the most beneficial element in this system, with stable cell pools and clones increasing antibody yields 6.5-fold and 6.75-fold respectively. Stability analysis demonstrated that the reduction in antibody expression, seen with cells transfected with the control vector over 120 generations, was mitigated in the clones containing A2UCOE-augmented transgenes. Analysis also showed that the A2UCOE reduced the amount of transgene promoter DNA methylation, which contributed to the maintenance of starting levels of expression.
Collapse
|
43
|
Müller-Kuller U, Ackermann M, Kolodziej S, Brendel C, Fritsch J, Lachmann N, Kunkel H, Lausen J, Schambach A, Moritz T, Grez M. A minimal ubiquitous chromatin opening element (UCOE) effectively prevents silencing of juxtaposed heterologous promoters by epigenetic remodeling in multipotent and pluripotent stem cells. Nucleic Acids Res 2015; 43:1577-92. [PMID: 25605798 PMCID: PMC4330381 DOI: 10.1093/nar/gkv019] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Epigenetic silencing of transgene expression represents a major obstacle for the efficient genetic modification of multipotent and pluripotent stem cells. We and others have demonstrated that a 1.5 kb methylation-free CpG island from the human HNRPA2B1-CBX3 housekeeping genes (A2UCOE) effectively prevents transgene silencing and variegation in cell lines, multipotent and pluripotent stem cells, and their differentiated progeny. However, the bidirectional promoter activity of this element may disturb expression of neighboring genes. Furthermore, the epigenetic basis underlying the anti-silencing effect of the UCOE on juxtaposed promoters has been only partially explored. In this study we removed the HNRPA2B1 moiety from the A2UCOE and demonstrate efficient anti-silencing properties also for a minimal 0.7 kb element containing merely the CBX3 promoter. This DNA element largely prevents silencing of viral and tissue-specific promoters in multipotent and pluripotent stem cells. The protective activity of CBX3 was associated with reduced promoter CpG-methylation, decreased levels of repressive and increased levels of active histone marks. Moreover, the anti-silencing effect of CBX3 was locally restricted and when linked to tissue-specific promoters did not activate transcription in off target cells. Thus, CBX3 is a highly attractive element for sustained, tissue-specific and copy-number dependent transgene expression in vitro and in vivo.
Collapse
Affiliation(s)
- Uta Müller-Kuller
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Hessen, 60596, Germany
| | - Mania Ackermann
- RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany Institute of Experimental Hematology, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany
| | - Stephan Kolodziej
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Hessen, 60596, Germany
| | - Christian Brendel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Hessen, 60596, Germany
| | - Jessica Fritsch
- RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany Institute of Experimental Hematology, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany
| | - Nico Lachmann
- RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany Institute of Experimental Hematology, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany
| | - Hana Kunkel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Hessen, 60596, Germany
| | - Jörn Lausen
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Hessen, 60596, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Moritz
- RG Reprogramming and Gene Therapy, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany Institute of Experimental Hematology, Hannover Medical School, Hannover, Lower Saxony, 30625, Germany
| | - Manuel Grez
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Hessen, 60596, Germany
| |
Collapse
|
44
|
Mao Y, Yan R, Li A, Zhang Y, Li J, Du H, Chen B, Wei W, Zhang Y, Sumners C, Zheng H, Li H. Lentiviral Vectors Mediate Long-Term and High Efficiency Transgene Expression in HEK 293T cells. Int J Med Sci 2015; 12:407-15. [PMID: 26005375 PMCID: PMC4441065 DOI: 10.7150/ijms.11270] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/20/2015] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVES Lentiviral vectors have been used successfully to rapidly produce decigram quantities of active recombinant proteins in mammalian cell lines. To optimize the protein production platform, the roles of Ubiquitous Chromatin Opening Element (UCOE), an insulator, and selected promoters were evaluated based on efficiency and stability of foreign gene expression mediated by lentiviral vectors. METHODS Five lentiviral vectors, pFIN-EF1α-GFP-2A-mCherH-WPRE containing EF1α promoter and HS4 insulator, p'HR.cppt.3'1.2kb-UCOE-SFFV-eGFP containing SFFV promoter and UCOE, pTYF-CMV(β-globin intron)-eGFP containing CMV promoter and β-globin intron, pTYF-CMV-eGFP containing CMV promoter, and pTYF-EF1α-eGFP with EF1α promoter were packaged, titered, and then transduced into 293T cells (1000 viral genomes per cell). The transduced cells were passaged once every three days at a ratio of 1:10. Expression level and stability of the foreign gene, green fluorescence protein (GFP), was evaluated using fluorescent microscopy and flow cytometry. Furthermore, we constructed a hepatitis C virus (HCV) E1 recombinant lentiviral vector, pLV-CMV-E1, driven by the CMV promoter. This vector was packaged and transduced into 293T cells, and the recombinant cell lines with stable expression of E1 protein were established by limiting dilution. RESULTS GFP expression in 293T cells transduced with the five lentiviral vectors peaked between passages 3 and 5 and persisted for more than 5 weeks. The expression was prolonged in the cells transduced with TYF-CMV (β-globin intron)-eGFP or TYF-CMV-eGFP, demonstrating less than a 50% decrease even at 9 weeks post transduction (p>0.05). The TYF-CMV-eGFP-transduced cells began with a higher level of GFP expression than other vectors did. The percentage of GFP positive cells for any of the five lentiviral vectors sustained over time. Moreover, the survival rates of all transfected cells exceeded 80% at both 5 and 9 weeks post transduction. Surprisingly, neither the HS4 insulator nor the UCOE sequence improved the GFP expression level or stability. Clonal cell lines with HCV E1 gene were generated from LV-CMV-E1 vector-infected 293T cells. A representative recombinant cell line maintained stable E1expression for at least 9 weeks without significant difference in morphology compared with untreated 293T cells. CONCLUSION The results suggest that all five vectors can stably transduce 293T cells, producing long term transgene expression with different efficiencies. However, neither the insulator nor the UCOE improved the GFP expression. The vectors containing the promoter CMV or CMV (β-globin intron) generated the highest gene expressions, manifesting as more favorable candidates for recombinant protein production in HEK293T cells.
Collapse
Affiliation(s)
- Yingying Mao
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Renhe Yan
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Andrew Li
- 2. Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yanling Zhang
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinlong Li
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyan Du
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Baihong Chen
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjin Wei
- 3. Beijing Minhai Biotechnology CO., LTD, Beijing, China
| | - Yi Zhang
- 4. Department of Pharmacology, University of Florida, Gainesville, Florida, USA
| | - Colin Sumners
- 5. Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Haifa Zheng
- 3. Beijing Minhai Biotechnology CO., LTD, Beijing, China
| | - Hongwei Li
- 1. School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Ramachandra DL, Shaw SSW, Shangaris P, Loukogeorgakis S, Guillot PV, Coppi PD, David AL. In utero therapy for congenital disorders using amniotic fluid stem cells. Front Pharmacol 2014; 5:270. [PMID: 25566071 PMCID: PMC4271591 DOI: 10.3389/fphar.2014.00270] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/18/2014] [Indexed: 12/15/2022] Open
Abstract
Congenital diseases are responsible for over a third of all pediatric hospital admissions. Advances in prenatal screening and molecular diagnosis have allowed the detection of many life-threatening genetic diseases early in gestation. In utero transplantation (IUT) with stem cells could cure affected fetuses but so far in humans, successful IUT using allogeneic hematopoietic stem cells (HSCs), has been limited to fetuses with severe immunologic defects and more recently IUT with allogeneic mesenchymal stem cell transplantation, has improved phenotype in osteogenesis imperfecta. The options of preemptive treatment of congenital diseases in utero by stem cell or gene therapy changes the perspective of congenital diseases since it may avoid the need for postnatal treatment and reduce future costs. Amniotic fluid stem (AFS) cells have been isolated and characterized in human, mice, rodents, rabbit, and sheep and are a potential source of cells for therapeutic applications in disorders for treatment prenatally or postnatally. Gene transfer to the cells with long-term transgenic protein expression is feasible. Recently, pre-clinical autologous transplantation of transduced cells has been achieved in fetal sheep using minimally invasive ultrasound guided injection techniques. Clinically relevant levels of transgenic protein were expressed in the blood of transplanted lambs for at least 6 months. The cells have also demonstrated the potential of repair in a range of pre-clinical disease models such as neurological disorders, tracheal repair, bladder injury, and diaphragmatic hernia repair in neonates or adults. These results have been encouraging, and bring personalized tissue engineering for prenatal treatment of genetic disorders closer to the clinic.
Collapse
Affiliation(s)
- Durrgah L. Ramachandra
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
| | - Steven S. W. Shaw
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Obstetrics and Gynaecology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Prenatal Therapy, Institute for Women’s Health, University College London, London, UK
| | - Panicos Shangaris
- Prenatal Therapy, Institute for Women’s Health, University College London, London, UK
| | - Stavros Loukogeorgakis
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
| | - Pascale V. Guillot
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
- Cellular Reprogramming and Perinatal Therapy, Institute for Women’s Health, University College London, London, UK
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
| | - Anna L. David
- Prenatal Therapy, Institute for Women’s Health, University College London, London, UK
| |
Collapse
|
46
|
Wong SP, Argyros O, Harbottle RP. Sustained expression from DNA vectors. ADVANCES IN GENETICS 2014; 89:113-152. [PMID: 25620010 DOI: 10.1016/bs.adgen.2014.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA vectors have the potential to become powerful medical tools for treatment of human disease. The human body has, however, developed a range of defensive strategies to detect and silence foreign or misplaced DNA, which is more typically encountered during infection or chromosomal damage. A clinically relevant human gene therapy vector must overcome or avoid these protections whilst delivering sustained levels of therapeutic gene product without compromising the vitality of the recipient host. Many non-viral DNA vectors trigger these defense mechanisms and are subsequently destroyed or rendered silent. Thus, without modification or considered design, the clinical utility of a typical DNA vector is fundamentally limited due to the transient nature of its transgene expression. The development of safe and persistently expressing DNA vectors is a crucial prerequisite for its successful clinical application and subsequently remains, therefore, one of the main strategic tasks of non-viral gene therapy research. In this chapter we will describe our current understanding of the mechanisms that can destroy or silence DNA vectors and discuss strategies, which have been utilized to improve their sustenance and the level and duration of their transgene expression.
Collapse
Affiliation(s)
- Suet Ping Wong
- Leukocyte Biology Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Orestis Argyros
- Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Richard P Harbottle
- DNA Vector Research, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
47
|
Dighe N, Khoury M, Mattar C, Chong M, Choolani M, Chen J, Antoniou MN, Chan JKY. Long-term reproducible expression in human fetal liver hematopoietic stem cells with a UCOE-based lentiviral vector. PLoS One 2014; 9:e104805. [PMID: 25118036 PMCID: PMC4130605 DOI: 10.1371/journal.pone.0104805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/14/2014] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic Stem Cell (HSC) targeted gene transfer is an attractive treatment option for a number of hematopoietic disorders caused by single gene defects. However, extensive methylation of promoter sequences results in silencing of therapeutic gene expression. The choice of an appropriate promoter is therefore crucial for reproducible, stable and long-term transgene expression in clinical gene therapy. Recent studies suggest efficient and stable expression of transgenes from the ubiquitous chromatin opening element (UCOE) derived from the human HNRPA2B1-CBX3 locus can be achieved in murine HSC. Here, we compared the use of HNRPA2B1-CBX3 UCOE (A2UCOE)-mediated transgene regulation to two other frequently used promoters namely EF1α and PGK in human fetal liver-derived HSC (hflHSC). Efficient transduction of hflHSC with a lentiviral vector containing an HNRPA2B1-CBX3 UCOE-eGFP (A2UCOE-eGFP) cassette was achieved at higher levels than that obtained with umbilical cord blood derived HSC (3.1x; p<0.001). While hflHSC were readily transduced with all three test vectors (A2UCOE-eGFP, PGK-eGFP and EF1α-eGFP), only the A2-UCOE construct demonstrated sustained transgene expression in vitro over 24 days (p<0.001). In contrast, within 10 days in culture a rapid decline in transgene expression in both PGK-eGFP and EF1α-eGFP transduced hflHSC was seen. Subsequently, injection of transduced cells into immunodeficient mice (NOD/SCID/Il2rg-/-) demonstrated sustained eGFP expression for the A2UCOE-eGFP group up to 10 months post transplantation whereas PGK-eGFP and EF1α-eGFP transduced hflHSC showed a 5.1 and 22.2 fold reduction respectively over the same time period. We conclude that the A2UCOE allows a more efficient and stable expression in hflHSC to be achieved than either the PGK or EF1α promoters and at lower vector copy number per cell.
Collapse
Affiliation(s)
- Niraja Dighe
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Maroun Khoury
- Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore
| | - Citra Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark Chong
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Mahesh Choolani
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianzhu Chen
- Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Michael N. Antoniou
- Department of Medical and Molecular Genetics, King's College London School of Medicine, Guys Hospital, London, United Kingdom
| | - Jerry K. Y. Chan
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Cancer and Stem Cell Program, Duke-NUS Graduate Medical School, Singapore, Singapore
- * E-mail:
| |
Collapse
|
48
|
Farinelli G, Capo V, Scaramuzza S, Aiuti A. Lentiviral vectors for the treatment of primary immunodeficiencies. J Inherit Metab Dis 2014; 37:525-33. [PMID: 24619149 DOI: 10.1007/s10545-014-9690-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 01/22/2023]
Abstract
In the last years important progress has been made in the treatment of several primary immunodeficiency disorders (PIDs) with gene therapy. Hematopoietic stem cell (HSC) gene therapy indeed represents a valid alternative to conventional transplantation when a compatible donor is not available and recent success confirmed the great potential of this approach. First clinical trials performed with gamma retroviral vectors were promising and guaranteed clinical benefits to the patients. On the other hand, the outcome of severe adverse events as the development of hematological abnormalities highlighted the necessity to develop a safer platform to deliver the therapeutic gene. Self-inactivating (SIN) lentiviral vectors (LVVs) were studied to overcome this hurdle through their preferable integration pattern into the host genome. In this review, we describe the recent advancements achieved both in vitro and at preclinical level with LVVs for the treatment of Wiskott-Aldrich syndrome (WAS), chronic granulomatous disease (CGD), ADA deficiency (ADA-SCID), Artemis deficiency, RAG1/2 deficiency, X-linked severe combined immunodeficiency (γchain deficiency, SCIDX1), X-linked lymphoproliferative disease (XLP) and immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome.
Collapse
Affiliation(s)
- Giada Farinelli
- Department of Pediatrics, Children's Hospital Bambino Gesù and University of Rome Tor Vergata School of Medicine, Rome, Italy
| | | | | | | |
Collapse
|
49
|
Abstract
Prostate cancer is very common in elderly men in developed countries. Unravelling the molecular and biological processes that contribute to tumor development and progressive growth, including its heterogeneity, is a challenging task. The fusion of the genes ERG and TMPRSS2 is the most frequent genomic alteration in prostate cancer. ERG is an oncogene that encodes a member of the family of ETS transcription factors. At lower frequency, other members of this gene family are also rearranged and overexpressed in prostate cancer. TMPRSS2 is an androgen-regulated gene that is preferentially expressed in the prostate. Most of the less frequent ETS fusion partners are also androgen-regulated and prostate-specific. During the last few years, novel concepts of the process of gene fusion have emerged, and initial experimental results explaining the function of the ETS genes ERG and ETV1 in prostate cancer have been published. In this review, we focus on the most relevant ETS gene fusions and summarize the current knowledge of the role of ETS transcription factors in prostate cancer. Finally, we discuss the clinical relevance of TMRPSS2-ERG and other ETS gene fusions in prostate cancer.
Collapse
Affiliation(s)
- Delila Gasi Tandefelt
- Departments of Pathology Urology, Erasmus University Medical Centre, PO Box 2040, 2000 CA Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
50
|
Lentiviral MGMT(P140K)-mediated in vivo selection employing a ubiquitous chromatin opening element (A2UCOE) linked to a cellular promoter. Biomaterials 2014; 35:7204-13. [PMID: 24875758 DOI: 10.1016/j.biomaterials.2014.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/01/2014] [Indexed: 12/17/2022]
Abstract
Notwithstanding recent successes, insertional mutagenesis as well as silencing and variegation of transgene expression still represent considerable obstacles to hematopoietic gene therapy. This also applies to O(6)-methylguanine DNA methyltransferase (MGMT)-mediated myeloprotection, a concept recently proven clinically effective in the context of glioblastoma therapy. To improve on this situation we here evaluate a SIN-lentiviral vector expressing the MGMT(P140K)-cDNA from a combined A2UCOE/PGK-promoter. In a murine in vivo chemoselection model the A2UCOE.PGK.MGMT construct allowed for significant myeloprotection as well as robust and stable selection of transgenic hematopoietic cells. In contrast, only transient enrichment and severe myelotoxicity was observed for a PGK.MGMT control vector. Selection of A2UCOE.PGK.MGMT-transduced myeloid and lymphoid mature and progenitor cells was demonstrated in the peripheral blood, bone marrow, spleen, and thymus. Unlike the PGK and SFFV promoters used as controls, the A2UCOE.PGK promoter allowed for sustained vector copy number-related transgene expression throughout the experiment indicating an increased resistance to silencing, which was further confirmed by CpG methylation studies of the PGK promoter. Thus, our data support a potential role of the A2UCOE.PGK.MGMT-vector in future MGMT-based myeloprotection and chemoselection strategies, and underlines the suitability of the A2UCOE element to stabilize lentiviral transgene expression in hematopoietic gene therapy.
Collapse
|