151
|
Luke GA, Ryan MD. Using the 2A Protein Coexpression System: Multicistronic 2A Vectors Expressing Gene(s) of Interest and Reporter Proteins. Methods Mol Biol 2018; 1755:31-48. [PMID: 29671261 DOI: 10.1007/978-1-4939-7724-6_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
To date, a huge range of different proteins-many with cotranslational and posttranslational subcellular localization signals-have been coexpressed together with various reporter proteins in vitro and in vivo using 2A peptides. The pros and cons of 2A co-expression technology are considered below, followed by a simple example of a "how to" protocol to concatenate multiple genes of interest, together with a reporter gene, into a single gene linked via 2As for easy identification or selection of transduced cells.
Collapse
Affiliation(s)
- Garry A Luke
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, Fife, Scotland, UK.
| | - Martin D Ryan
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, Fife, Scotland, UK
| |
Collapse
|
152
|
Petrovskaya LE, Shtefanyuk VS, Balaban PM, Ostrovsky MA, Malyshev AY. An analysis of the effect of the internal ribosome entry site of the encephalomyocarditis virus on the expression of the second gene in the bicistronic matrix in neurons of primary hippocampal cultures. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417040067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
153
|
Delivery of the gene encoding the tumor suppressor Sef into prostate tumors by therapeutic-ultrasound inhibits both tumor angiogenesis and growth. Sci Rep 2017; 7:15060. [PMID: 29118380 PMCID: PMC5678190 DOI: 10.1038/s41598-017-12408-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 08/30/2017] [Indexed: 11/16/2022] Open
Abstract
Carcinomas constitute over 80% of all human cancer types with no effective therapy for metastatic disease. Here, we demonstrate, for the first time, the efficacy of therapeutic-ultrasound (TUS) to deliver a human tumor suppressor gene, hSef-b, to prostate tumors in vivo. Sef is downregulated in various human carcinomas, in a manner correlating with tumor aggressiveness. In vitro, hSef-b inhibited proliferation of TRAMP C2 cells and attenuated activation of ERK/MAPK and the master transcription factor NF-κB in response to FGF and IL-1/TNF, respectively. In vivo, transfection efficiency of a plasmid co-expressing hSef-b/eGFP into TRAMP C2 tumors was 14.7 ± 2.5% following a single TUS application. Repeated TUS treatments with hSef-b plasmid, significantly suppressed prostate tumor growth (60%) through inhibition of cell proliferation (60%), and reduction in blood vessel density (56%). In accordance, repeated TUS-treatments with hSef-b significantly inhibited in vivo expression of FGF2 and MMP-9. FGF2 is a known mitogen, and both FGF2/MMP-9 are proangiogenic factors. Taken together our results strongly suggest that hSef-b acts in a cell autonomous as well as non-cell autonomous manner. Moreover, the study demonstrates the efficacy of non-viral TUS-based hSef-b gene delivery approach for the treatment of prostate cancer tumors, and possibly other carcinomas where Sef is downregulated.
Collapse
|
154
|
Wyatt A, Wartenberg P, Candlish M, Krasteva-Christ G, Flockerzi V, Boehm U. Genetic strategies to analyze primary TRP channel-expressing cells in mice. Cell Calcium 2017; 67:91-104. [DOI: 10.1016/j.ceca.2017.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 11/26/2022]
|
155
|
Kumagai H, Matsuura T, Kato Y, Watanabe H. Development of a bicistronic expression system in the branchiopod crustacean Daphnia magna. Genesis 2017; 55. [PMID: 29086479 DOI: 10.1002/dvg.23083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 01/07/2023]
Abstract
The viral 2A peptides have recently been used for bicistronic expression in various organisms. In this system, a single mRNA that codes for two proteins flanking the 2A peptide can be translated simultaneously into each protein by ribosomal skipping at this peptide sequence. Here, we tested the function of the Thosea asigna insect virus 2A (T2A) peptide in the branchiopod crustacean Daphnia magna-an emerging model of evolutionary developmental biology. First, we used transgenic Daphnia that expresses a potential bicistronic RNA containing mCherry and histone H2B- green fluorescent protein (GFP) open reading frames upstream and downstream of the T2A sequence, respectively. Microscopic observation revealed difference of localization of the two proteins in the cell, homogenous distribution of mCherry and nuclear localization of H2B-GFP. Second, we changed localization of mCherry from cytoplasm to plasma membrane by attachment of a consensus myristoylation motif in the bicistronic reporter. RNA that codes for this new bicistronic reporter was injected into eggs. At gastrulation stage, we found spectrally distinct fluorescence with enough intensity and resolution to detect membrane localized mCherry and nuclear GFP. These results indicate that the T2A peptide functions in D. magna and T2A-mediated bicistronic expression would be a promising tool for evo-devo studies of this species.
Collapse
Affiliation(s)
- Hitoshi Kumagai
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan
| | - Tomoaki Matsuura
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan
| | - Yasuhiko Kato
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan.,Frontier Research Base for Global Young Researchers, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan
| | - Hajime Watanabe
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
156
|
Eyvazi S, Kazemi B, Bandehpour M, Dastmalchi S. Identification of a novel single chain fragment variable antibody targeting CD24-expressing cancer cells. Immunol Lett 2017; 190:240-246. [DOI: 10.1016/j.imlet.2017.08.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/29/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
|
157
|
Nakabayashi H, Kawahara M, Nagamune T. Cell-Surface Expression Levels Are Important for Fine-Tuning the Performance of Receptor Tyrosine Kinase-Based Signalobodies. Biotechnol J 2017; 12. [PMID: 28881109 DOI: 10.1002/biot.201700441] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 08/18/2017] [Indexed: 11/07/2022]
Abstract
As receptor tyrosine kinases (RTKs) play important roles in cell-fate control of various cell types, engineered RTKs that could respond to inexpensive ligands might drastically reduce the cost of producing desired cells for various applications in regenerative medicine. We developed several engineered RTKs named "signalobodies" in which the ligand-recognition domain of RTKs is replaced by single-chain Fv for enabling recognition of a specific antigen. However, the remaining concern was the dysregulation of antigen-dependent on/off signaling of the signalobodies. This study aims at fine-tuning the performance of the signalobodies based on three RTKs (fibroblast growth factor receptor 1, insulin receptor, and c-fms). To this end, the cell-surface expression levels of the RTK-based signalobodies were altered by locating their genes either upstream or downstream of the internal ribosomal entry site, and by inserting 1 to 3 alanine residue(s) at the intracellular juxtamembrane region. As a result, while the signaling response was different among the three signalobodies, the antigen-dependent on/off regulation became tighter when the cell-surface expression levels of the signalobodies were lowered. Therefore, we successfully developed a method to diminish the leaky signaling of RTK-based signalobodies, which will be important for establishing the signalobody-based platform technology that can produce cells of interest for regenerative medicine.
Collapse
Affiliation(s)
- Hideto Nakabayashi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Masahiro Kawahara
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Teruyuki Nagamune
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
158
|
Saito S, Kurosawa A, Adachi N. Mechanistic basis for increased human gene targeting by promoterless vectors-roles of homology arms and Rad54 paralogs. FEBS J 2017. [DOI: 10.1111/febs.14137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Shinta Saito
- Graduate School of Nanobioscience; Yokohama City University; Japan
| | - Aya Kurosawa
- Graduate School of Nanobioscience; Yokohama City University; Japan
| | - Noritaka Adachi
- Graduate School of Nanobioscience; Yokohama City University; Japan
- Advanced Medical Research Center; Yokohama City University; Japan
| |
Collapse
|
159
|
Zhou X, Shi H, Yang S, Sun P. An efficient rapid system for assaying HBx-mediated transactivation. Biotechnol Lett 2017; 39:1091-1099. [PMID: 28386736 DOI: 10.1007/s10529-017-2334-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/30/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To develop a rapid and accurate assay system for screening inhibitors or enhancing agents targeting the transactivation capability of hepatitis B virus X protein (HBx) that activates cellular promoters in host cells to facilitate viral replication. RESULTS We constructed a new GFP-based reporter system which was different from a luciferase-based reporter system. Firstly, a FLAG-tagged HBx gene was inserted into an expression plasmid, resulting in plasmid pHBx. Next, HBx-FLAG was linked to EGFP by the internal ribosome entry site resulting in plasmid pHBxE. The transactivation effect of HBx-flag on cytomegalovirus (CMV) promoter was verified by EGFP expression using fluorescence quantitation and qPCR. Furthermore, the transactivation ability of the HBx gene was quantified by flow cytometry. Finally, this assay system was tested by known regulators of HBx including DDB1, ID1, and P53. As expected, the GFP reporter level in 293T cells changed with the increasing of HBx regulators. Furthermore, the system modeling the function of transactivation repressor in Hep3B, a HBV-integrated cell line. CONCLUSION Collectively, the GFP-based reporter system provides a rapid and accurate approach for analyzing transactivation ability of HBx.
Collapse
Affiliation(s)
- Xiaoling Zhou
- Stem Cell P2 Laboratory, Shantou University Medical College, Shantou, 515041, People's Republic of China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou, 515041, People's Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Haijun Shi
- Stem Cell P2 Laboratory, Shantou University Medical College, Shantou, 515041, People's Republic of China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou, 515041, People's Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Shaozhe Yang
- Stem Cell P2 Laboratory, Shantou University Medical College, Shantou, 515041, People's Republic of China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou, 515041, People's Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Pingnan Sun
- Stem Cell P2 Laboratory, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, People's Republic of China.
| |
Collapse
|
160
|
Novel insights into the expression pattern of anaphylatoxin receptors in mice and men. Mol Immunol 2017; 89:44-58. [PMID: 28600003 DOI: 10.1016/j.molimm.2017.05.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023]
Abstract
The anaphylatoxins (AT) C3a and C5a play important roles as mediators of inflammation. Further, they regulate and control multiple innate and adaptive immune responses through binding and activation of their cognate G protein-coupled receptors, i.e. C3a receptor (C3aR), C5a receptor 1 (C5aR1) and C5a receptor 2 (C5aR2), although the latter lacks important sequence motifs for G protein-coupling. Based on their pleiotropic functions, they contribute not only to tissue homeostasis but drive, perpetuate and resolve immune responses in many inflammatory diseases including infections, malignancies, autoimmune as well as allergic diseases. During the past few years, transcriptome expression data provided detailed insights into AT receptor tissue mRNA expression. In contrast, our understanding of cellular AT receptor expression in human and mouse tissues under steady and inflammatory conditions is still sketchy. Ligand binding studies, flow cytometric and immunohistochemical analyses convincingly demonstrated tissue-specific C5aR1 expression in various cells of myeloid origin. However, a detailed map for C3aR or C5aR2 expression in human or mouse tissue cells is still lacking. Also, reports about AT expression in lymphoid cells is still controversial. To understand the multiple roles of the ATs in the innate and adaptive immune networks, a detailed understanding of their receptor expression in health and disease is required. Recent findings obtained with novel GFP or tdTomato AT-receptor knock-in mice provide detailed insights into their expression pattern in tissue immune and stroma cells. Here, we will provide an update about our current knowledge of AT receptor expression pattern in humans and mice.
Collapse
|
161
|
Zhang B, Rapolu M, Kumar S, Gupta M, Liang Z, Han Z, Williams P, Su WW. Coordinated protein co-expression in plants by harnessing the synergy between an intein and a viral 2A peptide. PLANT BIOTECHNOLOGY JOURNAL 2017; 15:718-728. [PMID: 27879048 PMCID: PMC5425387 DOI: 10.1111/pbi.12670] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/16/2016] [Accepted: 11/19/2016] [Indexed: 05/22/2023]
Abstract
A novel approach is developed for coordinated expression of multiple proteins from a single transgene in plants. An Ssp DnaE mini-intein variant engineered for hyper-N-terminal autocleavage is covalently linked to the foot-and-mouth disease virus 2A (F2A) peptide with unique ribosome skipping property, via a peptide linker, to create an 'IntF2A' self-excising fusion protein domain. This IntF2A domain acts, in cis, to direct highly effective release of its flanking proteins of interest (POIs) from a 'polyprotein' precursor in plants. This is successfully demonstrated in stably transformed cultured tobacco cells as well as in different organs of transgenic tobacco plants. Highly efficient polyprotein processing mediated by the IntF2A domain was also demonstrated in lettuce and Nicotiana benthamiana based on transient expression. Protein constituents released from the polyprotein precursor displayed proper function and accumulated at similar levels inside the cells. Importantly, no C-terminal F2A extension remains on the released POIs. We demonstrated co-expression of as many as three proteins in plants without compromising expression levels when compared with those using single-protein vectors. Accurate differential cellular targeting of released POIs is also achieved. In addition, we succeeded in expressing a fully assembled and functional chimeric anti-His Tag antibody in N. benthamiana leaves. The IntF2A-based polyprotein transgene system overcomes key impediments of existing strategies for multiprotein co-expression in plants, which is particularly important for gene/trait stacking.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Molecular Biosciences and BioengineeringUniversity of Hawaii at ManoaHonoluluHIUSA
| | - Madhusudhan Rapolu
- Department of Molecular Biosciences and BioengineeringUniversity of Hawaii at ManoaHonoluluHIUSA
| | | | | | - Zhibin Liang
- Department of Molecular Biosciences and BioengineeringUniversity of Hawaii at ManoaHonoluluHIUSA
| | - Zhenlin Han
- Department of Molecular Biosciences and BioengineeringUniversity of Hawaii at ManoaHonoluluHIUSA
| | - Philip Williams
- Department of ChemistryUniversity of Hawaii at ManoaHonoluluHIUSA
| | - Wei Wen Su
- Department of Molecular Biosciences and BioengineeringUniversity of Hawaii at ManoaHonoluluHIUSA
| |
Collapse
|
162
|
TRPV1 channels are critical brain inflammation detectors and neuropathic pain biomarkers in mice. Nat Commun 2017; 8:15292. [PMID: 28489079 PMCID: PMC5436240 DOI: 10.1038/ncomms15292] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
The capsaicin receptor TRPV1 has been widely characterized in the sensory system as a key component of pain and inflammation. A large amount of evidence shows that TRPV1 is also functional in the brain although its role is still debated. Here we report that TRPV1 is highly expressed in microglial cells rather than neurons of the anterior cingulate cortex and other brain areas. We found that stimulation of microglial TRPV1 controls cortical microglia activation per se and indirectly enhances glutamatergic transmission in neurons by promoting extracellular microglial microvesicles shedding. Conversely, in the cortex of mice suffering from neuropathic pain, TRPV1 is also present in neurons affecting their intrinsic electrical properties and synaptic strength. Altogether, these findings identify brain TRPV1 as potential detector of harmful stimuli and a key player of microglia to neuron communication.
Collapse
|
163
|
Armbruster N, Krieg J, Weißenberger M, Scheller C, Steinert AF. Rescued Chondrogenesis of Mesenchymal Stem Cells under Interleukin 1 Challenge by Foamyviral Interleukin 1 Receptor Antagonist Gene Transfer. Front Pharmacol 2017; 8:255. [PMID: 28536528 PMCID: PMC5422547 DOI: 10.3389/fphar.2017.00255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/24/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) and their chondrogenic differentiation have been extensively investigated in vitro as MSCs provide an attractive source besides chondrocytes for cartilage repair therapies. Here we established prototype foamyviral vectors (FVV) that are derived from apathogenic parent viruses and are characterized by a broad host range and a favorable integration pattern into the cellular genome. As the inflammatory cytokine interleukin 1 beta (IL1β) is frequently present in diseased joints, the protective effects of FVV expressing the human interleukin 1 receptor antagonist protein (IL1RA) were studied in an established in vitro model (aggregate culture system) of chondrogenesis in the presence of IL1β. Materials and Methods: We generated different recombinant FVVs encoding enhanced green fluorescent protein (EGFP) or IL1RA and examined their transduction efficiencies and transgene expression profiles using different cell lines and human primary MSCs derived from bone marrow-aspirates. Transgene expression was evaluated by fluorescence microscopy (EGFP), flow cytometry (EGFP), and ELISA (IL1RA). For evaluation of the functionality of the IL1RA transgene to block the inhibitory effects of IL1β on chondrogenesis of primary MSCs and an immortalized MSC cell line (TERT4 cells), the cells were maintained following transduction as aggregate cultures in standard chondrogenic media in the presence or absence of IL1β. After 3 weeks of culture, pellets were harvested and analyzed by histology and immunohistochemistry for chondrogenic phenotypes. Results: The different FVV efficiently transduced cell lines as well as primary MSCs, thereby reaching high transgene expression levels in 6-well plates with levels of around 100 ng/ml IL1RA. MSC aggregate cultures which were maintained in chondrogenic media without IL1β supplementation revealed a chondrogenic phenotype by means of strong positive staining for collagen type II and matrix proteoglycan (Alcian blue). Addition of IL1β was inhibitory to chondrogenesis in untreated control pellets. In contrast, foamyviral mediated IL1RA expression rescued the chondrogenesis in pellets cultured in the presence of IL1β. Transduced MSC pellets reached thereby very high IL1RA transgene expression levels with a peak of 1087 ng/ml after day 7, followed by a decrease to 194 ng/ml after day 21, while IL1RA concentrations of controls were permanently below 200 pg/ml. Conclusion: Our results indicate that FVV are capable of efficient gene transfer to MSCs, while reaching IL1RA transgene expression levels, that were able to efficiently block the impacts of IL1β in vitro. FVV merit further investigation as a means to study the potential as a gene transfer tool for MSC based therapies for cartilage repair.
Collapse
Affiliation(s)
- Nicole Armbruster
- Institute for Virology and Immunobiology, University of WuerzburgWuerzburg, Germany.,Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| | - Jennifer Krieg
- Institute for Virology and Immunobiology, University of WuerzburgWuerzburg, Germany.,Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| | - Manuel Weißenberger
- Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| | - Carsten Scheller
- Institute for Virology and Immunobiology, University of WuerzburgWuerzburg, Germany
| | - Andre F Steinert
- Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| |
Collapse
|
164
|
Yang C, Shang X, Cheng L, Yang L, Liu X, Bai C, Wei Z, Hua J, Li G. DNMT 1 maintains hypermethylation of CAG promoter specific region and prevents expression of exogenous gene in fat-1 transgenic sheep. PLoS One 2017; 12:e0171442. [PMID: 28158319 PMCID: PMC5291418 DOI: 10.1371/journal.pone.0171442] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/20/2017] [Indexed: 02/06/2023] Open
Abstract
Methylation is an important issue in gene expression regulation and also in the fields of genetics and reproduction. In this study, we created fat-1 transgenic sheep, investigated the fine-mapping and the modulatory mechanisms of promoter methylation. Sheep fetal fibroblasts were transfected by pCAG-fat1-IRES-EGFP. Monoclonal cell line was screened as nuclear donor and carried out nuclear transfer (441 transgenic cloned embryos, 52 synchronism recipient sheep). Six offsprings were obtained. Expressions of exogenous genes fat-1 and EGFP were detectable in 10 examined tissues and upregulated omega-3 fatty acid content. Interestingly, more or less EGFP negative cells were detectable in the positive transgenic fetal skin cells. EGFP negative and positive cells were sorted by flow cytometry, and their methylation status in the whole promoter region (1701 nt) were investigated by bisulphate sequencing. The fine-mapping of methylation in CAG promoter were proposed. The results suggested that exogenous gene expression was determined by the methylation status from 721–1346 nt and modulated by methylation levels at 101, 108 and 115 nt sites in CAG promoter. To clarify the regulatory mechanism of methylation, examination of four DNA methyltransferases (DNMTs) demonstrated that hypermethylation of CAG promoter is mainly maintained by DNMT 1 in EGFP negative cells. Furthermore, investigation of the cell surface antigen CD34, CD45 and CD166 indicated that EGFP positive and negative cells belong to different types. The present study systematically clarified methylation status of CAG promoter in transgenic sheep and regulatory mechanism, which will provide research strategies for gene expression regulation in transgenic animals.
Collapse
Affiliation(s)
- Chunrong Yang
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xueying Shang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Lei Cheng
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
| | - Lei Yang
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
| | - Xuefei Liu
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
| | - Chunling Bai
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
| | - Zhuying Wei
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
| | - Jinlian Hua
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Guangpeng Li
- Research Center for Laboratory Animal Science, Inner Mongolia University, Hohhot, China
- * E-mail:
| |
Collapse
|
165
|
Kaiser GG, Mucci NC, González V, Sánchez L, Parrón JA, Pérez MD, Calvo M, Aller JF, Hozbor FA, Mutto AA. Detection of recombinant human lactoferrin and lysozyme produced in a bitransgenic cow. J Dairy Sci 2017; 100:1605-1617. [PMID: 28109583 DOI: 10.3168/jds.2016-11173] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 10/07/2016] [Indexed: 02/02/2023]
Abstract
Lactoferrin and lysozyme are 2 glycoproteins with great antimicrobial activity, being part of the nonspecific defensive system of human milk, though their use in commercial products is difficult because human milk is a limited source. Therefore, many investigations have been carried out to produce those proteins in biological systems, such as bacteria, yeasts, or plants. Mammals seem to be more suitable as expression systems for human proteins, however, especially for those that are glycosylated. In the present study, we developed a bicistronic commercial vector containing a goat β-casein promoter and an internal ribosome entry site fragment between the human lactoferrin and human lysozyme genes to allow the introduction of both genes into bovine adult fibroblasts in a single transfection. Embryos were obtained by somatic cell nuclear transfer, and, after 6 transferences to recipients, 3 pregnancies and 1 viable bitransgenic calf were obtained. The presence of the vector was confirmed by fluorescent in situ hybridization of skin cells. At 13 mo of life and after artificial induction of lactation, both recombinant proteins were found in the colostrum and milk of the bitransgenic calf. Human lactoferrin concentration in the colostrum was 0.0098 mg/mL and that in milk was 0.011 mg/mL; human lysozyme concentration in the colostrum was 0.0022 mg/mL and that in milk was 0.0024 mg/mL. The molar concentration of both human proteins revealed no differences in protein production of the internal ribosome entry site upstream and downstream protein. The enzymatic activity of lysozyme in the transgenic milk was comparable to that of human milk, being 6 and 10 times higher than that of bovine lysozyme present in milk. This work represents an important step to obtain multiple proteins or enhance single protein production by using animal pharming and fewer regulatory and antibiotic-resistant foreign sequences, allowing the design of humanized milk with added biological value for newborn nutrition and development. Transgenic animals can offer a unique opportunity to the dairy industry, providing starting materials suitable to develop specific products with high added value.
Collapse
Affiliation(s)
- Germán G Kaiser
- Grupo de Biotecnología de la Reproducción, Instituto Nacional de Tecnología Agropecuaria, 7620 Balcarce, Argentina.
| | - Nicolás C Mucci
- Grupo de Biotecnología de la Reproducción, Instituto Nacional de Tecnología Agropecuaria, 7620 Balcarce, Argentina
| | - Vega González
- Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Lourdes Sánchez
- Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - José A Parrón
- Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - María D Pérez
- Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Miguel Calvo
- Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Juan F Aller
- Grupo de Biotecnología de la Reproducción, Instituto Nacional de Tecnología Agropecuaria, 7620 Balcarce, Argentina
| | - Federico A Hozbor
- Grupo de Biotecnología de la Reproducción, Instituto Nacional de Tecnología Agropecuaria, 7620 Balcarce, Argentina
| | - Adrián A Mutto
- Laboratorio Biotecnologías Aplicadas a la Reproducción y Mejoramiento Genético Animal, Instituto de Investigaciones Biotechnològicas-Instituto Tecnològico Chascomùs (IIB-INTECH), Universidad Nacional de San Martin-Consejo de Investigaciones Cientìficas y Tècnicas (CONICET), 1650 San Martin, Argentina
| |
Collapse
|
166
|
Significant changes in endogenous retinal gene expression assessed 1 year after a single intraocular injection of AAV-CNTF or AAV-BDNF. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16078. [PMID: 27933306 PMCID: PMC5142514 DOI: 10.1038/mtm.2016.78] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/27/2016] [Accepted: 10/12/2016] [Indexed: 12/17/2022]
Abstract
Use of viral vectors to deliver therapeutic genes to the central nervous system holds promise for the treatment of neurodegenerative diseases and neurotrauma. Adeno-associated viral (AAV) vectors encoding brain-derived neurotrophic factor (BDNF) or ciliary derived neurotrophic factor (CNTF) promote the viability and regeneration of injured adult rat retinal ganglion cells. However, these growth-inducing transgenes are driven by a constitutively active promoter, thus we examined whether long-term AAV-mediated secretion of BDNF or CNTF affected endogenous retinal gene expression. One year after the intravitreal injection of AAV-green fluorescent protein (GFP), bi-cistronic AAV-BDNF-GFP or AAV-CNTF-GFP, mRNA was extracted and analyzed using custom 96 well polymerase chain reaction arrays. Of 93 test genes, 56% showed significantly altered expression in AAV-BDNF-GFP and/or AAV-CNTF-GFP retinas compared with AAV-GFP controls. Of these genes, 73% showed differential expression in AAV-BDNF versus AAV-CNTF injected eyes. To focus on retinal ganglion cell changes, quantitative polymerase chain reaction was undertaken on mRNA (16 genes) obtained from fixed retinal sections in which the ganglion cell layer was enriched. The sign and extent of fold changes in ganglion cell layer gene expression differed markedly from whole retinal samples. Sustained and global alteration in endogenous mRNA expression after gene therapy should be factored into any interpretation of experimental/clinical outcomes, particularly when introducing factors into the central nervous system that require secretion to evoke functionality.
Collapse
|
167
|
Liu Y, Li K, Gao Y, Gao L, Zhong L, Zhang Y, Liu C, Zhang Y, Wang X. Recombinant Marek's Disease Virus as a Vector-Based Vaccine against Avian Leukosis Virus Subgroup J in Chicken. Viruses 2016; 8:v8110301. [PMID: 27827933 PMCID: PMC5127015 DOI: 10.3390/v8110301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 10/18/2016] [Accepted: 10/26/2016] [Indexed: 11/21/2022] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) is an immunosuppressive virus that causes considerable economic losses to the chicken industry in China. However, there is currently no effective vaccine to prevent ALV-J infection. In order to reduce the losses caused by ALV-J, we constructed two effective ALV-J vaccines by inserting the ALV-J (strain JL093-1) env or gag+env genes into the US2 gene of the Marek’s disease herpesviruses (MDV) by transfection of overlapping fosmid DNAs, creating two recombinant MDVs, rMDV/ALV-gag+env and rMDV/ALV-env. Analysis of cultured chicken embryo fibroblasts infected with the rMDVs revealed that Env and Gag were successfully expressed and that there was no difference in growth kinetics in cells infected with rMDVs compared with that of cells infected with the parent MDV. Chickens vaccinated with either rMDV revealed that positive serum antibodies were induced. Both rMDVs also effectively reduced the rate of positive viremia in chicken flocks challenged with ALV-J. The protective effect provided by rMDV/ALV-env inoculation was slightly stronger than that provided by rMDV/ALV-gag+env. This represents the first study where a potential rMDV vaccine, expressing ALV-J antigenic genes, has been shown to be effective in the prevention of ALV-J. Our study also opens new avenues for the control of MDV and ALV-J co-infection.
Collapse
Affiliation(s)
- Yongzhen Liu
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Kai Li
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yulong Gao
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Li Gao
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Li Zhong
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yao Zhang
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Changjun Liu
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yanping Zhang
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Xiaomei Wang
- Avian Immunosuppressive Diseases Division, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| |
Collapse
|
168
|
Wood L, Booth DG, Vargiu G, Ohta S, deLima Alves F, Samejima K, Fukagawa T, Rappsilber J, Earnshaw WC. Auxin/AID versus conventional knockouts: distinguishing the roles of CENP-T/W in mitotic kinetochore assembly and stability. Open Biol 2016; 6:150230. [PMID: 26791246 PMCID: PMC4736828 DOI: 10.1098/rsob.150230] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Most studies using knockout technologies to examine protein function have relied either on shutting off transcription (conventional conditional knockouts with tetracycline-regulated gene expression or gene disruption) or destroying the mature mRNA (RNAi technology). In both cases, the target protein is lost at a rate determined by its intrinsic half-life. Thus, protein levels typically fall over at least 1-3 days, and cells continue to cycle while exposed to a decreasing concentration of the protein. Here we characterise the kinetochore proteome of mitotic chromosomes isolated from a cell line in which the essential kinetochore protein CENP-T is present as an auxin-inducible degron (AID) fusion protein that is fully functional and able to support the viability of the cells. Stripping of the protein from chromosomes in early mitosis via targeted proteasomal degradation reveals the dependency of other proteins on CENP-T for their maintenance in kinetochores. We compare these results with the kinetochore proteome of conventional CENP-T/W knockouts. As the cell cycle is mostly formed from G1, S and G2 phases a gradual loss of CENP-T/W levels is more likely to reflect dependencies associated with kinetochore assembly pre-mitosis and upon entry into mitosis. Interestingly, a putative super-complex involving Rod-Zw10-zwilch (RZZ complex), Spindly, Mad1/Mad2 and CENP-E requires the function of CENP-T/W during kinetochore assembly for its stable association with the outer kinetochore, but once assembled remains associated with chromosomes after stripping of CENP-T during mitosis. This study highlights the different roles core kinetochore components may play in the assembly of kinetochores (upon entry into mitosis) versus the maintenance of specific components (during mitosis).
Collapse
Affiliation(s)
- Laura Wood
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| | - Daniel G Booth
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| | - Giulia Vargiu
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| | - Shinya Ohta
- Center for Innovative and Translational Medicine, Kochi University, Kochi, Japan
| | - Flavia deLima Alves
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| | - Kumiko Samejima
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Suita 565-0871, Japan
| | - Juri Rappsilber
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK Institute of Bioanalytics, Department of Biotechnology, Technische Universität Berlin, Berlin 13353, Germany
| | - William C Earnshaw
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Michael Swann Building, Kings Buildings, Mayfield Road, Edinburgh EH9 3JR, UK
| |
Collapse
|
169
|
Zhu K, Liu D, Lai H, Li J, Wang C. Developing miRNA therapeutics for cardiac repair in ischemic heart disease. J Thorac Dis 2016; 8:E918-E927. [PMID: 27747027 DOI: 10.21037/jtd.2016.08.93] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) families have been found to be powerful regulators in a wide variety of diseases, which enables the possible use of miRNAs in therapeutic strategies for cardiac repair after ischemic heart disease. This review provides some general insights into miRNAs modulation for development of current molecular and cellular therapeutics in cardiac repair, including endogenous regeneration, endogenous repair, stem cells transplantation, and reprogramming. We also review the delivery strategies for miRNAs modulation, and briefly summarize the current bench and clinical efforts that are being made to explore miRNAs as the future therapeutic target.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;; Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Dingqian Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;; Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;; Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;; Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;; Shanghai Institute of Cardiovascular Disease, Shanghai 200032, China
| |
Collapse
|
170
|
Fowler DK, Stewart S, Seredick S, Eisen JS, Stankunas K, Washbourne P. A MultiSite Gateway Toolkit for Rapid Cloning of Vertebrate Expression Constructs with Diverse Research Applications. PLoS One 2016; 11:e0159277. [PMID: 27500400 PMCID: PMC4976983 DOI: 10.1371/journal.pone.0159277] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/29/2016] [Indexed: 11/19/2022] Open
Abstract
Recombination-based cloning is a quick and efficient way to generate expression vectors. Recent advancements have provided powerful recombinant DNA methods for molecular manipulations. Here, we describe a novel collection of three-fragment MultiSite Gateway cloning system-compatible vectors providing expanded molecular tools for vertebrate research. The components of this toolkit encompass a broad range of uses such as fluorescent imaging, dual gene expression, RNA interference, tandem affinity purification, chemically-inducible dimerization and lentiviral production. We demonstrate examples highlighting the utility of this toolkit for producing multi-component vertebrate expression vectors with diverse primary research applications. The vectors presented here are compatible with other Gateway toolkits and collections, facilitating the rapid generation of a broad range of innovative DNA constructs for biological research.
Collapse
Affiliation(s)
- Daniel K. Fowler
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Scott Stewart
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Steve Seredick
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Judith S. Eisen
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Kryn Stankunas
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Philip Washbourne
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| |
Collapse
|
171
|
Reimer EN, Walenda G, Seidel E, Scholl UI. CACNA1H(M1549V) Mutant Calcium Channel Causes Autonomous Aldosterone Production in HAC15 Cells and Is Inhibited by Mibefradil. Endocrinology 2016; 157:3016-22. [PMID: 27258646 DOI: 10.1210/en.2016-1170] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We recently demonstrated that a recurrent gain-of-function mutation in a T-type calcium channel, CACNA1H(M1549V), causes a novel Mendelian disorder featuring early-onset primary aldosteronism and hypertension. This variant was found independently in five families. CACNA1H(M1549V) leads to impaired channel inactivation and activation at more hyperpolarized potentials, inferred to cause increased calcium entry. We here aimed to study the effect of this variant on aldosterone production. We heterologously expressed empty vector, CACNA1H(WT) and CACNA1H(M1549V) in the aldosterone-producing adrenocortical cancer cell line H295R and its subclone HAC15. Transfection rates, expression levels, and subcellular distribution of the channel were similar between CACNA1H(WT) and CACNA1H(M1549V). We measured aldosterone production by an ELISA and CYP11B2 (aldosterone synthase) expression by real-time PCR. In unstimulated cells, transfection of CACNA1H(WT) led to a 2-fold increase in aldosterone levels compared with vector-transfected cells. Expression of CACNA1H(M1549V) caused a 7-fold increase in aldosterone levels. Treatment with angiotensin II or increased extracellular potassium levels further stimulated aldosterone production in both CACNA1H(WT)- and CACNA1H(M1549V)-transfected cells. Similar results were obtained for CYP11B2 expression. Inhibition of CACNA1H channels with the T-type calcium channel blocker Mibefradil completely abrogated the effects of CACNA1H(WT) and CACNA1H(M1549V) on CYP11B2 expression. These results directly link CACNA1H(M1549V) to increased aldosterone production. They suggest that calcium channel blockers may be beneficial in the treatment of a subset of patients with primary aldosteronism. Such blockers could target CACNA1H or both CACNA1H and the L-type calcium channel CACNA1D that is also expressed in the adrenal gland and mutated in patients with primary aldosteronism.
Collapse
Affiliation(s)
- Esther N Reimer
- Department of Nephrology, Medical School, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Gudrun Walenda
- Department of Nephrology, Medical School, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Eric Seidel
- Department of Nephrology, Medical School, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Ute I Scholl
- Department of Nephrology, Medical School, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
172
|
Stewart HJ, Ralph GS, Fong-Wong L, Strickland I, McCloskey L, Barnes L, Blount I, Wells O, Truran CJM, Kingsman AJ, Palfi S, Mitrophanous KA. Optimizing Transgene Configuration and Protein Fusions to Maximize Dopamine Production for the Gene Therapy of Parkinson's Disease. HUM GENE THER CL DEV 2016; 27:100-10. [PMID: 27470285 DOI: 10.1089/humc.2016.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pharmacological dopamine replacement therapies provide the most well-established treatments for Parkinson's disease (PD). However, these long-term treatments can lead to motor complications and off-target effects. ProSavin(®), a lentiviral vector (LV)-based gene therapy approach aimed at restoring local and continuous dopamine production, through delivery of three enzymes in the dopamine biosynthesis pathway, was demonstrated to be safe and well-tolerated in a phase I/II clinical study of patients with advanced PD. Although improvements in motor behaviour were observed, the data indicated that higher levels of dopamine replacement might be required to maximize benefit. We attempted to increase production of dopamine, and its precursor L-Dopa in LV-transduced cells, by optimizing the gene order in the ProSavin expression cassette, and by creating fusions of two or three of the transgenes, using linker sequences. In vitro analysis showed that several gene arrangements provided significantly increased dopamine and/or L-Dopa production compared with ProSavin, and that LV titers and transgene expression were not affected by introducing gene fusions. One vector, equine infectious anemia virus (EIAV)-TCiA, was selected for further characterization and showed significant improvements in dopamine and L-Dopa production compared with ProSavin, in human neuronal cells. Further characterization of EIAV-TCiA demonstrated expression of all three dopamine enzymes in vivo and faithful delivery and integration of the expected gene expression cassette within the genome of target cells, as assessed by Northern and Southern blotting. In conclusion, we have developed a novel LV vector with an increased capacity for L-Dopa and dopamine production compared with the current ProSavin vector. Clinical evaluation of this vector will be performed to assess the benefits in patients with PD.
Collapse
Affiliation(s)
| | - G Scott Ralph
- 1 Oxford BioMedica (UK) Ltd. , Oxford, United Kingdom
| | - Liang Fong-Wong
- 2 Henry Wellcome L.I.N.E., University of Bristol , Bristol BS1 3NY, United Kingdom
| | - Iain Strickland
- 2 Henry Wellcome L.I.N.E., University of Bristol , Bristol BS1 3NY, United Kingdom
| | | | - Lucy Barnes
- 1 Oxford BioMedica (UK) Ltd. , Oxford, United Kingdom
| | - Ian Blount
- 1 Oxford BioMedica (UK) Ltd. , Oxford, United Kingdom
| | - Owen Wells
- 1 Oxford BioMedica (UK) Ltd. , Oxford, United Kingdom
| | | | | | - Stéphane Palfi
- 3 AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, Neurochirurgie, INSERM IMRB U955 eq.14, Université Paris Est Créteil (UPEC) , Faculté de Médecine, Creteil, F-94010, France
| | | |
Collapse
|
173
|
Li W, Yap MW, Voss V, Stoye JP. Expression levels of Fv1: effects on retroviral restriction specificities. Retrovirology 2016; 13:42. [PMID: 27342974 PMCID: PMC4921018 DOI: 10.1186/s12977-016-0276-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/16/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mouse protein Fv1 is a factor that can confer resistance to retroviral infection. The two major Fv1 alleles from laboratory mice, Fv1 (n) and Fv1 (b) , restrict infection by different murine leukaemia viruses (MLVs). Fv1(n) restricts B-tropic MLV, but not N-tropic MLV or NB-tropic MLV. In cells expressing Fv1(b) at natural levels, only N-MLV is restricted, however restriction of NB-MLV and partial restriction of B-MLV were observed when recombinant Fv1(b) was expressed from an MLV promoter in Fv1 null Mus dunni tail fibroblast cells. To investigate the relationship between expression level and restriction specificity we have developed new retroviral delivery vectors which allow inducible expression of Fv1, and yet allow sufficient production of fluorescent reporter proteins for analysis in our FACS-based restriction assay. RESULTS We demonstrated that at concentrations close to the endogenous expression level, Fv1(b) specifically restricts only N-MLV, but restriction of NB-MLV, and to a lesser extent B-MLV, could be gained by increasing the protein level of Fv1(b). By contrast, we found that even when Fv1(n) is expressed at very high levels, no significant inhibition of N-MLV or NB-MLV could be observed. Study of Fv1 mutants using this assay led to the identification of determinants for N/B tropism at an expression level close to that of endogenous Fv1(n) and Fv1(b). We also compared the recently described restriction activities of wild mice Fv1 proteins directed against non-MLV retroviruses when expressed at different levels. Fv1 from M. spretus restricted N-MLV, B-MLV and equine infectious anaemia virus equally even at low concentrations, while Fv1 from M. macedonicus showed even stronger restriction against equine infectious anaemia virus than to N-MLV. Restriction of feline foamy virus by Fv1 of M. caroli occurred at levels equivalent to MLV restriction. CONCLUSIONS Our data indicate that for some but not all Fv1 proteins, gain of restriction activities could be achieved by increasing the expression level of Fv1. However such a concentration dependent effect is not seen with most Fv1s and cannot explain the recently reported activities against non-MLVs. It will be interesting to examine whether overexpression of other capsid binding restriction factors such as TRIM5α or Mx2 result in novel restriction specificities.
Collapse
Affiliation(s)
- Wilson Li
- Retrovirus-Host Interactions Laboratory, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Melvyn W Yap
- Retrovirus-Host Interactions Laboratory, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Vicky Voss
- Retrovirus-Host Interactions Laboratory, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Jonathan P Stoye
- Retrovirus-Host Interactions Laboratory, The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK. .,Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
174
|
Fuchs SP, Martinez-Navio JM, Gao G, Desrosiers RC. Recombinant AAV Vectors for Enhanced Expression of Authentic IgG. PLoS One 2016; 11:e0158009. [PMID: 27332822 PMCID: PMC4917256 DOI: 10.1371/journal.pone.0158009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022] Open
Abstract
Adeno-associated virus (AAV) has become a vector of choice for the treatment of a variety of genetic diseases that require safe and long-term delivery of a missing protein. Muscle-directed gene transfer for delivery of protective antibodies against AIDS viruses and other pathogens has been used experimentally in mice and monkeys. Here we examined a number of variations to AAV vector design for the ability to produce authentic immunoglobulin G (IgG) molecules. Expression of rhesus IgG from a single single-stranded AAV (ssAAV) vector (one vector approach) was compared to expression from two self-complementary AAV (scAAV) vectors, one for heavy chain and one for light chain (two vector approach). Both the one vector and the two vector approaches yielded considerable levels of expressed full-length IgG. A number of modifications to the ssAAV expression system were then examined for their ability to increase the efficiency of IgG expression. Inclusion of a furin cleavage sequence with a linker peptide just upstream of the 2A self-cleaving sequence from foot-and-mouth disease virus (F2A) increased IgG expression approximately 2 fold. Inclusion of these sequences also helped to ensure a proper sequence at the C-terminal end of the heavy chain. Inclusion of the post-transcriptional regulatory element from woodchuck hepatitis virus (WPRE) further increased IgG expression 1.5–2.0 fold. IgG1 versions of the two rhesus IgGs that were examined consistently expressed better than the IgG2 forms. In contrast to what has been reported for AAV2-mediated expression of other proteins, introduction of capsid mutations Y445F and Y731F did not increase ssAAV1-mediated expression of IgG as determined by transduction experiments in cell culture. Our findings provide a rational basis for AAV vector design for expression of authentic IgG.
Collapse
Affiliation(s)
- Sebastian P. Fuchs
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - José M. Martinez-Navio
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
175
|
Cesaratto F, Burrone OR, Petris G. Tobacco Etch Virus protease: A shortcut across biotechnologies. J Biotechnol 2016; 231:239-249. [PMID: 27312702 DOI: 10.1016/j.jbiotec.2016.06.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/31/2016] [Accepted: 06/10/2016] [Indexed: 11/29/2022]
Abstract
About thirty years ago, studies on the RNA genome of Tobacco Etch Virus revealed the presence of an efficient and specific protease, called Tobacco Etch Virus protease (TEVp), that was part of the Nuclear Inclusion a (NIa) enzyme. TEVp is an efficient and specific protease of 27kDa that has become a valuable biotechnological tool. Nowadays TEVp is a unique endopeptidase largely exploited in biotechnology from industrial applications to in vitro and in vivo cellular studies. A number of TEVp mutants with different rate of cleavage, stability and specificity have been reported. Similarly, a panel of different target cleavage sites, derived from the canonical ENLYFQ-G/S site, has been established. In this review we describe these aspects of TEVp and some of its multiple applications. A particular focus is on the use and molecular biology of TEVp in living cells and organisms.
Collapse
Affiliation(s)
- Francesca Cesaratto
- International Centre for Genetic Engineering and Biotechnology, ICGEB, Trieste, Italy
| | - Oscar R Burrone
- International Centre for Genetic Engineering and Biotechnology, ICGEB, Trieste, Italy.
| | | |
Collapse
|
176
|
Roellecke K, Virts EL, Einholz R, Edson KZ, Altvater B, Rossig C, von Laer D, Scheckenbach K, Wagenmann M, Reinhardt D, Kramm CM, Rettie AE, Wiek C, Hanenberg H. Optimized human CYP4B1 in combination with the alkylator prodrug 4-ipomeanol serves as a novel suicide gene system for adoptive T-cell therapies. Gene Ther 2016; 23:615-26. [PMID: 27092941 DOI: 10.1038/gt.2016.38] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/23/2016] [Accepted: 04/05/2016] [Indexed: 12/18/2022]
Abstract
Engineering autologous or allogeneic T cells to express a suicide gene can control potential toxicity in adoptive T-cell therapies. We recently reported the development of a novel human suicide gene system that is based on an orphan human cytochrome P450 enzyme, CYP4B1, and the naturally occurring alkylator prodrug 4-ipomeanol. The goal of this study was to systematically develop a clinically applicable self-inactivating lentiviral vector for efficient co-expression of CYP4B1 as an ER-located protein with two distinct types of cell surface proteins, either MACS selection genes for donor lymphocyte infusions after allogeneic stem cell transplantation or chimeric antigen receptors for retargeting primary T cells. The U3 region of the myeloproliferative sarcoma virus in combination with the T2A site was found to drive high-level expression of our CYP4B1 mutant with truncated CD34 or CD271 as MACS suitable selection markers. This lentiviral vector backbone was also well suited for co-expression of CYP4B1 with a codon-optimized CD19 chimeric antigen receptor (CAR) construct. Finally, 4-ipomeanol efficiently induced apoptosis in primary T cells that co-express mutant CYP4B1 and the divergently located MACS selection and CAR genes. In conclusion, we here developed a clinically suited lentiviral vector that supports high-level co-expression of cell surface proteins with a potent novel human suicide gene.
Collapse
Affiliation(s)
- K Roellecke
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - E L Virts
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Einholz
- Institute for Organic Chemistry, University of Tübingen, Tübingen, Germany
| | - K Z Edson
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - B Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - C Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - D von Laer
- Institute for Virology, Innsbruck Medical University, Innsbruck, Austria
| | - K Scheckenbach
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - M Wagenmann
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - D Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - C M Kramm
- Division of Pediatric Hematology and Oncology, Department of Child and Adolescent Health, University Medical Center Göttingen, Göttingen, Germany
| | - A E Rettie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - C Wiek
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - H Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
177
|
Halstead JM, Wilbertz JH, Wippich F, Lionnet T, Ephrussi A, Chao JA. TRICK: A Single-Molecule Method for Imaging the First Round of Translation in Living Cells and Animals. Methods Enzymol 2016; 572:123-57. [PMID: 27241753 DOI: 10.1016/bs.mie.2016.02.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
The life of an mRNA is dynamic within a cell. The development of quantitative fluorescent microscopy techniques to image single molecules of RNA has allowed many aspects of the mRNA lifecycle to be directly observed in living cells. Recent advances in live-cell multicolor RNA imaging, however, have now made it possible to investigate RNA metabolism in greater detail. In this chapter, we present an overview of the design and implementation of the translating RNA imaging by coat protein knockoff RNA biosensor, which allows untranslated mRNAs to be distinguished from ones that have undergone a round of translation. The methods required for establishing this system in mammalian cell lines and Drosophila melanogaster oocytes are described here, but the principles may be applied to any experimental system.
Collapse
Affiliation(s)
- J M Halstead
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - J H Wilbertz
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - F Wippich
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - T Lionnet
- Transcription Imaging Consortium, HHMI Janelia Research Campus, Ashburn, VA, United States
| | - A Ephrussi
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - J A Chao
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
178
|
Wilmington SR, Matouschek A. An Inducible System for Rapid Degradation of Specific Cellular Proteins Using Proteasome Adaptors. PLoS One 2016; 11:e0152679. [PMID: 27043013 PMCID: PMC4820223 DOI: 10.1371/journal.pone.0152679] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/17/2016] [Indexed: 11/25/2022] Open
Abstract
A common way to study protein function is to deplete the protein of interest from cells and observe the response. Traditional methods involve disrupting gene expression but these techniques are only effective against newly synthesized proteins and leave previously existing and stable proteins untouched. Here, we introduce a technique that induces the rapid degradation of specific proteins in mammalian cells by shuttling the proteins to the proteasome for degradation in a ubiquitin-independent manner. We present two implementations of the system in human culture cells that can be used individually to control protein concentration. Our study presents a simple, robust, and flexible technology platform for manipulating intracellular protein levels.
Collapse
Affiliation(s)
- Shameika R. Wilmington
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
| | - Andreas Matouschek
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States of America
- * E-mail:
| |
Collapse
|
179
|
Park A, Yun T, Hill TE, Ikegami T, Juelich TL, Smith JK, Zhang L, Freiberg AN, Lee B. Optimized P2A for reporter gene insertion into Nipah virus results in efficient ribosomal skipping and wild-type lethality. J Gen Virol 2016; 97:839-843. [PMID: 26781134 PMCID: PMC4854364 DOI: 10.1099/jgv.0.000405] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/15/2016] [Indexed: 11/18/2022] Open
Abstract
Incorporation of reporter genes within virus genomes is an indispensable tool for interrogation of virus biology and pathogenesis. In previous work, we incorporated a fluorophore into a viral ORF by attaching it to the viral gene via a P2A ribosomal skipping sequence. This recombinant Nipah virus, however, was attenuated in vitro relative to WT virus. In this work, we determined that inefficient ribosomal skipping was a major contributing factor to this attenuation. Inserting a GSG linker before the P2A sequence resulted in essentially complete skipping, significantly improved growth in vitro, and WT lethality in vivo. To the best of our knowledge, this represents the first time a recombinant virus of Mononegavirales with integration of a reporter into a viral ORF has been compared with the WT virus in vivo. Incorporating the GSG linker for improved skipping efficiency whenever functionally important is a critical consideration for recombinant virus design.
Collapse
Affiliation(s)
- Arnold Park
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tatyana Yun
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Terence E. Hill
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Tetsuro Ikegami
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Terry L. Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennifer K. Smith
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lihong Zhang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Benhur Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
180
|
Kusabuka H, Fujiwara K, Tokunaga Y, Hirobe S, Nakagawa S, Okada N. Highly efficient gene transfer using a retroviral vector into murine T cells for preclinical chimeric antigen receptor-expressing T cell therapy. Biochem Biophys Res Commun 2016; 473:73-79. [PMID: 26993168 DOI: 10.1016/j.bbrc.2016.03.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
Abstract
Adoptive immunotherapy using chimeric antigen receptor-expressing T (CAR-T) cells has attracted attention as an efficacious strategy for cancer treatment. To prove the efficacy and safety of CAR-T cell therapy, the elucidation of immunological mechanisms underlying it in mice is required. Although a retroviral vector (Rv) is mainly used for the introduction of CAR to murine T cells, gene transduction efficiency is generally less than 50%. The low transduction efficiency causes poor precision in the functional analysis of CAR-T cells. We attempted to improve the Rv gene transduction protocol to more efficiently generate functional CAR-T cells by optimizing the period of pre-cultivation and antibody stimulation. In the improved protocol, gene transduction efficiency to murine T cells was more than 90%. In addition, almost all of the prepared murine T cells expressed CAR after puromycin selection. These CAR-T cells had antigen-specific cytotoxic activity and secreted multiple cytokines by antigen stimulation. We believe that our optimized gene transduction protocol for murine T cells contributes to the advancement of T cell biology and development of immunotherapy using genetically engineered T cells.
Collapse
Affiliation(s)
- Hotaka Kusabuka
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kento Fujiwara
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yusuke Tokunaga
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sachiko Hirobe
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shinsaku Nakagawa
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Naoki Okada
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
181
|
Lesueur LL, Mir LM, André FM. Overcoming the Specific Toxicity of Large Plasmids Electrotransfer in Primary Cells In Vitro. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e291. [PMID: 27111417 PMCID: PMC5014460 DOI: 10.1038/mtna.2016.4] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 12/23/2015] [Indexed: 11/10/2022]
Abstract
Gene electrotransfer is a safe and efficient nonviral technique for the transfer of nucleic acids of all sizes. Using a small reporter plasmid (3.5 kbp), electrotransfer of more than 90% of the cells, with ~70% viability, can be routinely achieved even in primary cells like mesenchymal stem cells. However, under the same experimental conditions, electrotransfer of larger plasmids (from 6 to 16 kbp) results in very low viability and transfection efficacy. Here, we show that these strong decreases are directly linked to the physical size of the plasmid molecule. Moreover, large plasmids are toxic only when the cells are exposed to electrotransfer pulses. This specific toxicity of large plasmids during electrotransfer is not due to transgene expression and occurs within less than 45 minutes. Indeed, postpulses recovery times of up to 45 minutes are able to entirely abolish the specific toxicity of large plasmid electrotransfer, resulting in a survival and transfection efficacy identical to that of small plasmids. Finally, electrotransfer of small and large plasmids can reach 90–99% of transfection with 60–90% survival considering the findings here reported.
Collapse
Affiliation(s)
- Léa L Lesueur
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Lluis M Mir
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Franck M André
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
182
|
Vivek Srinivas VM, Basagoudanavar SH, Hosamani M. IRES mediated expression of viral 3C protease for enhancing the yield of FMDV empty capsids using baculovirus system. Biologicals 2016; 44:64-8. [PMID: 26775685 DOI: 10.1016/j.biologicals.2015.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/23/2015] [Accepted: 12/04/2015] [Indexed: 11/25/2022] Open
Abstract
For expression of FMDV empty capsids, high protease activity associated with 3C co-expressed with P1 polyprotein has been reported to adversely affect the yields of capsids. Limiting the levels of 3Cpro relative to P1-2A polypeptide is thus critical to enhance the yields. In this study, FMDV internal ribosome entry site (IRES) sequence which serves as an alternative to the CAP-dependent translation initiation mechanism, was used for controlled translation of 3C protease. Baculovirus expressing bicistronic cDNA cassette containing two open reading frames-FMDV capsid gene (P1-2A) and 3Cpro intervened by IRES was prepared. Analysis of the expression in insect cells infected with baculovirus showed increased accumulation of processed capsids. Recombinant capsids showed higher immunoreactivity similar to the whole virus antigen, when reacted with polyclonal antibodies against the purified whole virus 146S particles. Thus, inclusion of the IRES upstream of 3Cpro facilitated reduced expression of the protease in baculovirus expression system, without causing significant proteolysis, thereby contributing to improved yields of the processed capsid antigens.
Collapse
Affiliation(s)
- V M Vivek Srinivas
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore 560 024, India
| | | | - Madhusudan Hosamani
- ICAR-Indian Veterinary Research Institute, Hebbal, Bangalore 560 024, India.
| |
Collapse
|
183
|
Tseng AWS, Chen C, Breslin MB, Lan MS. Tumor-specific promoter-driven adenoviral therapy for insulinoma. Cell Oncol (Dordr) 2016; 39:279-86. [PMID: 26902080 DOI: 10.1007/s13402-016-0274-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Insulinomas are the most common type of neuroendocrine (NE) pancreatic islet tumors. Patients with insulinomas may develop complications associated with hyperinsulinemia. To increase the treatment options for insulinoma patients, we have tested a conditionally replicating adenovirus that has been engineered in such a way that it can specifically express therapeutic genes in NE tumors. METHODS We used a promoter-specific adenoviral vector delivery system that is regulated by an INSM1 (insulinoma-associated-1) promoter, which is silent in normal adult tissues but active in developing NE cells and tumors. Through a series of modifications, using an insulator (HS4) and neuron-restrictive silencer elements (NRSEs), an oncolytic adenoviral vector was generated that retains tumor specificity and drives the expression of a mutated adenovirus E1A gene (Δ24E1A) and the herpes simplex virus thymidine kinase (HSV-tk) gene. The efficacy of this vector was tested in insulinoma-derived MIN, RIN, βTC-1 and pancreatic (Panc-1) cells using in vitro cell survival and in vivo tumor growth assays. RESULTS Using in vitro insulinoma-derived cell lines and an in vivo subcutaneous mouse tumor model we found that the INSM1 promoter-driven viruses were able to replicate specifically in INSM1-positive cells. INSM1-specific HSV-tk expression in combination with ganciclovir treatment resulted in dose-dependent tumor cell killing, leaving INSM1-negative cells unharmed. When we combined the INSM1-promoter driven HSV-tk with Δ24E1A and INSM1p-HSV-tk (K5) viruses, we found that the co-infected insulinoma-derived cells expressed higher levels of HSV-tk and exhibited more efficient tumor suppression than cells infected with INSM1p-HSV-tk virus alone. CONCLUSIONS INSM1 promoter-driven conditionally replicating adenoviruses may serve as a new tool for the treatment of insulinoma and may provide clinicians with additional options to combat this disease.
Collapse
Affiliation(s)
- Alan Wei-Shun Tseng
- The Research Institute for Children, Children's Hospital, 200 Henry Clay Avenue, New Orleans, LA, 70118, USA.,Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Chiachen Chen
- The Research Institute for Children, Children's Hospital, 200 Henry Clay Avenue, New Orleans, LA, 70118, USA.,Laboratory of Diana Helis Henry Medical Research Foundation, New Orleans, LA, 70119, USA
| | - Mary B Breslin
- The Research Institute for Children, Children's Hospital, 200 Henry Clay Avenue, New Orleans, LA, 70118, USA.,Laboratory of Diana Helis Henry Medical Research Foundation, New Orleans, LA, 70119, USA.,Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Michael S Lan
- The Research Institute for Children, Children's Hospital, 200 Henry Clay Avenue, New Orleans, LA, 70118, USA. .,Laboratory of Diana Helis Henry Medical Research Foundation, New Orleans, LA, 70119, USA. .,Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA. .,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
184
|
Jung U, Takahashi M, Rossi JJ, Burnett JC. LGIT In Vitro Latency Model in Primary and T Cell Lines to Test HIV-1 Reactivation Compounds. Methods Mol Biol 2016; 1354:255-264. [PMID: 26714717 DOI: 10.1007/978-1-4939-3046-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Persistent latent HIV-1 reservoirs pose a major barrier for combinatorial antiretroviral therapy (cART) to achieve eradication of the virus. A variety of mechanisms likely contribute to HIV-1 persistence, including establishment of post-integration latency in resting CD4+ T-lymphocytes, the proliferation of these latently infected cells, and the induced or spontaneous reactivation of latent virus. To elucidate the mechanisms of latency and to investigate therapeutic strategies for reactivating and purging the latent reservoir, investigators have developed in vitro models of HIV-1 latency using primary CD4+ T-lymphocytes and CD4+ T-cell lines. Several types of in vitro latency models range from replication-competent to single-round, replication-deficient viruses exhibiting different degrees of viral genomic deletion. Working under the hypothesis that HIV-1 post-integration latency is directly linked to HIV-1 promoter activity, which can be obscured by additional proteins expressed during replication, we focus here on the creation of latently infected primary human T-cells and cell lines through the single-round, replication deficient HIV-1 LGIT model. In this model the long terminal repeat (LTR) of the HIV-1 virus drives a cassette of GFP-IRES-Tat that allows testing of reactivating components and initiates a positive feedback loop through Tat expression.
Collapse
Affiliation(s)
- Ulrike Jung
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Mayumi Takahashi
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - John J Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Fox North, 1001A, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - John C Burnett
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Fox North, 1001A, 1500 Duarte Rd., Duarte, CA, 91010, USA.
| |
Collapse
|
185
|
Brennig S, Lachmann N, Buchegger T, Hetzel M, Schambach A, Moritz T. Chemoprotection of murine hematopoietic cells by combined gene transfer of cytidine deaminase (CDD) and multidrug resistance 1 gene (MDR1). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:148. [PMID: 26651614 PMCID: PMC4676838 DOI: 10.1186/s13046-015-0260-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/16/2015] [Indexed: 01/23/2023]
Abstract
Background Hematologic toxicity represents a major side effect of cytotoxic chemotherapy frequently preventing adequately dosed chemotherapy application and impeding therapeutic success. Transgenic (over)expression of chemotherapy resistance (CTX-R) genes in hematopoietic stem- and progenitor cells represents a potential strategy to overcome this problem. To apply this concept in the context of acute myeloid leukemia and myelodysplasia, we have investigated the overexpression of the multidrug resistance 1 (MDR1) and the cytidine deaminase (CDD) gene conferring resistance to anthracyclines and cytarabine (Ara-C), the two most important drugs in the treatment of these diseases. Methods State-of-the-art, third generation, self-inactivating (SIN) lentiviral vectors were utilized to overexpress a human CDD-cDNA and a codon-optimized human MDR1-cDNA corrected for cryptic splice sites from a spleen focus forming virus derived internal promoter. Studies were performed in myeloid 32D cells as well as primary lineage marker negative (lin−) murine bone marrow cells and flow cytometric analysis of suspension cultures and clonogenic analysis of vector transduced cells following cytotoxic drug challenge were utilized as read outs. Results Efficient chemoprotection of CDD and MDR1 transduced hematopoietic 32D as well as primary lin− cells was proven in the context of Ara-C and anthracycline application. Both, CTX-R transduced 32D as well as primary hematopoietic cells displayed marked resistance at concentrations 5–20 times the LD50 of non-transduced control cells. Moreover, simultaneous CDD/MDR1 gene transfer resulted in similar protection levels even when combined Ara-C anthracycline treatment was applied. Furthermore, significant enrichment of transduced cells was observed upon cytotoxic drug administration. Conclusions Our data demonstrate efficient chemoprotection as well as enrichment of transduced cells in hematopoietic cell lines as well as primary murine hematopoietic progenitor cells following Ara-C and/or anthracycline application, arguing for the efficacy as well as feasibility of our approach and warranting further evaluation of this concept. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0260-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Brennig
- Reprogramming and Gene Therapy Group, REBIRTH Cluster-of Excellence, Hannover Medical School, Carl-Neuberg-Str.1, Hannover, D-30625, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- Reprogramming and Gene Therapy Group, REBIRTH Cluster-of Excellence, Hannover Medical School, Carl-Neuberg-Str.1, Hannover, D-30625, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,JRG Translational Hematology of Congenital Diseases, REBIRTH Cluster-of Excellence, Hannover Medical School, Hannover, Germany
| | - Theresa Buchegger
- Reprogramming and Gene Therapy Group, REBIRTH Cluster-of Excellence, Hannover Medical School, Carl-Neuberg-Str.1, Hannover, D-30625, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Miriam Hetzel
- Reprogramming and Gene Therapy Group, REBIRTH Cluster-of Excellence, Hannover Medical School, Carl-Neuberg-Str.1, Hannover, D-30625, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Thomas Moritz
- Reprogramming and Gene Therapy Group, REBIRTH Cluster-of Excellence, Hannover Medical School, Carl-Neuberg-Str.1, Hannover, D-30625, Germany. .,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
186
|
Telpalo-Carpio SA, Diaz-Mitoma F, Moreno-Cuevas JE, Aguilar-Yáñez JM. Internal ribosome entry site (IRES) from Encephalomyocarditis virus (EMCV) as a tool for shuttle expression plasmids. Biochem Biophys Res Commun 2015; 468:548-53. [DOI: 10.1016/j.bbrc.2015.10.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 11/28/2022]
|
187
|
Wiszniak S, Scherer M, Ramshaw H, Schwarz Q. Neuropilin-2 genomic elements drive cre recombinase expression in primitive blood, vascular and neuronal lineages. Genesis 2015; 53:709-17. [PMID: 26454009 DOI: 10.1002/dvg.22905] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/28/2015] [Accepted: 10/07/2015] [Indexed: 12/23/2022]
Abstract
We have established a novel Cre mouse line, using genomic elements encompassing the Nrp2 locus, present within a bacterial artificial chromosome clone. By crossing this Cre driver line to R26R LacZ reporter mice, we have documented the temporal expression and lineage traced tissues in which Cre is expressed. Nrp2-Cre drives expression in primitive blood cells arising from the yolk sac, venous and lymphatic endothelial cells, peripheral sensory ganglia, and the lung bud. This mouse line will provide a new tool to researchers wishing to study the development of various tissues and organs in which this Cre driver is expressed, as well as allow tissue-specific knockout of genes of interest to study protein function. This work also presents the first evidence for expression of Nrp2 protein in a mesodermal progenitor with restricted hematopoietic potential, which will significantly advance the study of primitive erythropoiesis. genesis 53:709-717, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sophie Wiszniak
- Centre for Cancer Biology and University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
| | - Michaela Scherer
- Centre for Cancer Biology and University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
| | - Hayley Ramshaw
- Centre for Cancer Biology and University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology and University of South Australia, Frome Road, Adelaide, South Australia, 5000, Australia
| |
Collapse
|
188
|
Den Hartogh SC, Passier R. Concise Review: Fluorescent Reporters in Human Pluripotent Stem Cells: Contributions to Cardiac Differentiation and Their Applications in Cardiac Disease and Toxicity. Stem Cells 2015; 34:13-26. [DOI: 10.1002/stem.2196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 07/14/2015] [Accepted: 07/28/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Sabine C. Den Hartogh
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
- Department of Applied Stem cell Technologies. MIRA Institute for Biomedical Technology and Technical Medicine; University of Twente, P.O.Box 217; Enschede The Netherlands
| |
Collapse
|
189
|
Candlish M, Angelis RD, Götz V, Boehm U. Gene Targeting in Neuroendocrinology. Compr Physiol 2015; 5:1645-76. [DOI: 10.1002/cphy.c140079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
190
|
Recombinant bovine adenovirus-3 co-expressing bovine respiratory syncytial virus glycoprotein G and truncated glycoprotein gD of bovine herpesvirus-1 induce immune responses in cotton rats. Mol Biotechnol 2015; 57:58-64. [PMID: 25173687 DOI: 10.1007/s12033-014-9801-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
One of the impediments in the development of safe and cost effective vaccines for veterinary use has been the availability of appropriate delivery vehicle. We have chosen to develop and use bovine adenovirus (BAdV)-3 as vaccine delivery vector in cattle. Here, we describe the construction of recombinant E3 deleted BAdV-3 vectors expressing single vaccine antigen (BAV360; bovine respiratory syncytial virus G) or two vaccine antigens (BAV851; bovine herpesvirus-1gDt and bovine respiratory syncytial virus G). Recombinant proteins expressed by BAV360 or BAV851 were recognized by protein-specific monoclonal antibodies. Moreover, intranasal immunization of cotton rats with BAV360 (expressing a single vaccine antigen) or BAV851 (expressing two vaccine antigens) induced strong antigen-specific immune responses. These results suggest that single replication-competent BAdV-3 expressing vaccine antigens of two economically important respiratory pathogens of calves has potential to act as a feasible approach in the development of economically effective veterinary vaccines for cattle.
Collapse
|
191
|
WANG J, HU Y, TAN BQ, WANG JJ, ZHAO MT, WENG QJ, ZHU DF, WANG HY. [Construction of expression vector pLCK-CD69-IRES-EGFP and generation of CD69 transgenic mice]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2015; 44:511-516. [PMID: 26713525 PMCID: PMC10397002 DOI: 10.3785/j.issn.1008-9292.2015.09.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/20/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To construct the expression vector pLCK-CD69-IRES-EGFP that contains mouse cell surface activation protein CD69 and enhanced green fluorescent protein(EGFP),and to generate CD69 transgenic mice based on this vector. METHODS First, RNA was extracted from mouse lung tissue and cDNA was synthesized via reverse transcription. PCR primer was designed through the PubMed searching, then mouse CD69 DNA fragment was amplified with PCR. Second, this DNA fragment was subcloned to the pInsulater-LCK-IRES-EGFP plasmid and constructed the transgenic vector after the verification of nucleotide sequence. Third, the expression vector was then transfected into 293 T cells and its expression in 293 T cells was observed under fluorescence microscope. Last, microinjection was performed to transfer the expression vector pLCK-CD69-IRES-EGFP into fertilized eggs, which were implanted into pseudo-pregnant recipient mice. After birth the tail samples of the pups were obtained for the purpose of genotyping to determine the transgenic founders. Fluorescence microscope and flow cytometer were used to measure the expression of CD69 on cells. RESULTS The construction of the expression vector pLCK-CD69-IRES-EGFP was verified by enzyme digestion and DNA sequencing. The transfected 293 T cell showed expression of the protein under fluorescence microscope. Identification of PCR for the tail tissue of the pups confirmed the present of CD69 transgene and resting lymphocytes demonstrated the expression of CD69. CONCLUSION The construction of expression vector pLCK-CD69-IRES-EGFP and generation of CD69 transgenic mice have been successfully processed, which lays a foundation of the solid pattern studies in inflammatory diseases.
Collapse
|
192
|
A light-switchable bidirectional expression module allowing simultaneous regulation of multiple genes. Biochem Biophys Res Commun 2015; 465:769-76. [PMID: 26301633 DOI: 10.1016/j.bbrc.2015.08.085] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/20/2015] [Indexed: 11/23/2022]
Abstract
Several light-regulated genetic circuits have been applied to spatiotemporally control transgene expression in mammalian cells. However, simultaneous regulation of multiple genes using one genetic device by light has not yet been reported. In this study, we engineered a bidirectional expression module based on LightOn system. Our data showed that both reporter genes could be regulated at defined and quantitative levels. Simultaneous regulation of four genes was further achieved in cultured cells and mice. Additionally, we successfully utilized the bidirectional expression module to monitor the expression of a suicide gene, showing potential for photodynamic gene therapy. Collectively, we provide a robust and useful tool to simultaneously control multiple genes expression by light, which will be widely used in biomedical research and biotechnology.
Collapse
|
193
|
Lang A, Neuhaus J, Pfeiffenberger M, Schröder E, Ponomarev I, Weber Y, Gaber T, Schmidt MFG. Optimization of a nonviral transfection system to evaluate Cox-2 controlled interleukin-4 expression for osteoarthritis gene therapy in vitro. J Gene Med 2015; 16:352-63. [PMID: 25382123 DOI: 10.1002/jgm.2812] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 06/15/2014] [Accepted: 11/03/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Gene therapy appears to have the potential for achieving a long-term remedy for osteoarthritis (OA). However, there is a risk of adverse reactions, especially when using cytomegalovirus-controlled expression. To provide a safe application, we focused on the expression of therapeutic cytokines [e.g. interleukin (IL)-4] in a disease-responsive manner by use of the previously cloned Cox-2 promoter as 'genetic switch'. In the present study, we report the functionality of a controlled gene therapeutic system in an equine osteoarthritic cell model. METHODS Different nonviral transfection reagents were tested for their efficiency on equine chondrocytes stimulated with equine IL-1β or lipopolysaccharide to create an inflammatory environment. To optimize the transfection, we successfully redesigned the vector by excluding the internal ribosomal entry site (IRES). The functionality of our Cox-2 promoter construct with respect to expressing IL-4 was proven at the mRNA and protein levels and the anti-inflammatory potential of IL-4 was confirmed by analyzing the expression of IL-1β, IL-6, IL-8, matrix metalloproteinase (MMP)-1, MMP-3 and tumor necrosis factor (TNF)-α using a quantitative polymerase chain reaction. RESULTS Nonviral transfection reagents yielded transfection rates from 21% to 44% with control vectors with and without IRES, respectively. Stimulation of equine chondrocytes resulted in a 20-fold increase of mRNA expression of IL-1β. Such exogenous stimulation of chondrocytes transfected with pNCox2-IL4 led to an increase of IL-4 mRNA expression, whereas expression of inflammatory mediators decreased. The timely link between these events confirms the anti-inflammatory potential of synthesized IL-4. CONCLUSIONS We consider that this approach has significant potential for translation into a useful anti-inflammation therapy. Molecular tools such as the described therapeutic plasmid pave the way for a local-controlled, self-limiting gene therapy.
Collapse
Affiliation(s)
- Annemarie Lang
- Institute of Immunology, Department of Veterinary Medicine, Freie Universität Berlin, Germany; Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany; German Rheumatism Research Center, Berlin, Germany; Berlin-Brandenburg School of Regenerative Therapies, Charité University Hospital, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Abstract
The human heart has a limited capacity to regenerate lost or damaged cardiomyocytes after cardiac insult. Instead, myocardial injury is characterized by extensive cardiac remodeling by fibroblasts, resulting in the eventual deterioration of cardiac structure and function. Cardiac function would be improved if these fibroblasts could be converted into cardiomyocytes. MicroRNAs (miRNAs), small noncoding RNAs that promote mRNA degradation and inhibit mRNA translation, have been shown to be important in cardiac development. Using this information, various researchers have used miRNAs to promote the formation of cardiomyocytes through several approaches. Several miRNAs acting in combination promote the direct conversion of cardiac fibroblasts into cardiomyocytes. Moreover, several miRNAs have been identified that aid the formation of inducible pluripotent stem cells and miRNAs also induce these cells to adopt a cardiac fate. MiRNAs have also been implicated in resident cardiac progenitor cell differentiation. In this review, we discuss the current literature as it pertains to these processes, as well as discussing the therapeutic implications of these findings.
Collapse
Affiliation(s)
- Conrad P Hodgkinson
- From the Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Martin H Kang
- From the Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Sophie Dal-Pra
- From the Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Maria Mirotsou
- From the Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Victor J Dzau
- From the Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Department of Medicine, Duke University Medical Center, Durham, NC.
| |
Collapse
|
195
|
Hoekstra ME, Dijkgraaf FE, Schumacher TN, Rohr JC. Assessing T lymphocyte function and differentiation by genetically encoded reporter systems. Trends Immunol 2015; 36:392-400. [DOI: 10.1016/j.it.2015.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/15/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
|
196
|
Lachmann N, Brennig S, Hillje R, Schermeier H, Phaltane R, Dahlmann J, Gruh I, Heinz N, Schiedlmeier B, Baum C, Moritz T. Tightly regulated 'all-in-one' lentiviral vectors for protection of human hematopoietic cells from anticancer chemotherapy. Gene Ther 2015; 22:883-92. [PMID: 26125609 DOI: 10.1038/gt.2015.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 06/09/2015] [Accepted: 06/18/2015] [Indexed: 01/12/2023]
Abstract
Successful application of gene therapy strategies may require stringently regulated transgene expression. Along this line, we describe a doxycycline (Dox)-inducible 'all-in-one' lentiviral vector design using the pTET-T11 (TII) minimal-promoter and a reverse transactivator protein (rtTA2S-M2) driven by the phosphoglycerate kinase promoter allowing for tight regulation of transgene expression (Lv.TII vectors). Vector design was evaluated in human hematopoietic cells in the context of cytidine deaminase (hCDD)-based myeloprotective gene therapy. Upon Dox administration, a rapid (16-24 h) and dose-dependent (>0.04 μg ml(-1) Dox) onset of transgene expression was detected in Lv.TII.CDD gene-modified K562 cells as well as in primary human CD34(+) hematopoietic cells. Importantly, in both cell models low background transgene expression was observed in the absence of Dox. Functionality of Dox-inducible hCDD expression was demonstrated by >10-fold increase in cytosine arabinoside (1-β-d-arabinofuranosylcytosine, Ara-C) resistance of Lv.TII.CDD-transduced K562 cells. In addition, Lv.TII.CDD-transduced CD34(+)-derived myeloid cells were protected from up to 300 nm Ara-C (control affected from 50 nm onwards). These data clearly demonstrate the suitability of our self-inactivating lentiviral vector to induce robust, tightly regulated transgene expression in human hematopoietic cells with minimal background activity and highlight the potential of our construct in myeloprotective gene therapy strategies.
Collapse
Affiliation(s)
- N Lachmann
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - S Brennig
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - R Hillje
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - H Schermeier
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - R Phaltane
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - J Dahlmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - I Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - N Heinz
- LOEWE-Research Group for (targeted) Gene Modification in Stem Cells, Paul-Ehrlich-Institute, Langen, Germany
| | - B Schiedlmeier
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - C Baum
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - T Moritz
- Reprogramming and Gene Therapy Group, REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
197
|
Guerrero AD, Moyes JS, Cooper LJN. The human application of gene therapy to re-program T-cell specificity using chimeric antigen receptors. CHINESE JOURNAL OF CANCER 2015; 33:421-33. [PMID: 25189715 PMCID: PMC4190432 DOI: 10.5732/cjc.014.10100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The adoptive transfer of T cells is a promising approach to treat cancers. Primary human T cells can be modified using viral and non-viral vectors to promote the specific targeting of cancer cells via the introduction of exogenous T-cell receptors (TCRs) or chimeric antigen receptors (CARs). This gene transfer displays the potential to increase the specificity and potency of the anticancer response while decreasing the systemic adverse effects that arise from conventional treatments that target both cancerous and healthy cells. This review highlights the generation of clinical-grade T cells expressing CARs for immunotherapy, the use of these cells to target B-cell malignancies and, particularly, the first clinical trials deploying the Sleeping Beauty gene transfer system, which engineers T cells to target CD19+ leukemia and non-Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Alan D Guerrero
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
198
|
Viral bimolecular fluorescence complementation: a novel tool to study intracellular vesicular trafficking pathways. PLoS One 2015; 10:e0125619. [PMID: 25915798 PMCID: PMC4411132 DOI: 10.1371/journal.pone.0125619] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/24/2015] [Indexed: 01/23/2023] Open
Abstract
The Human Immunodeficiency Virus type 1 (HIV-1) accessory protein Nef interacts with a multitude of cellular proteins, manipulating the host membrane trafficking machinery to evade immune surveillance. Nef interactions have been analyzed using various in vitro assays, co-immunoprecipitation studies, and more recently mass spectrometry. However, these methods do not evaluate Nef interactions in the context of viral infection nor do they define the sub-cellular location of these interactions. In this report, we describe a novel bimolecular fluorescence complementation (BiFC) lentiviral expression tool, termed viral BiFC, to study Nef interactions with host cellular proteins in the context of viral infection. Using the F2A cleavage site from the foot and mouth disease virus we generated a viral BiFC expression vector capable of concurrent expression of Nef and host cellular proteins; PACS-1, MHC-I and SNX18. Our studies confirmed the interaction between Nef and PACS-1, a host membrane trafficking protein involved in Nef-mediated immune evasion, and demonstrated co-localization of this complex with LAMP-1 positive endolysosomal vesicles. Furthermore, we utilized viral BiFC to localize the Nef/MHC-I interaction to an AP-1 positive endosomal compartment. Finally, viral BiFC was observed between Nef and the membrane trafficking regulator SNX18. This novel demonstration of an association between Nef and SNX18 was localized to AP-1 positive vesicles. In summary, viral BiFC is a unique tool designed to analyze the interaction between Nef and host cellular proteins by mapping the sub-cellular locations of their interactions during viral infection.
Collapse
|
199
|
Nikić I, Kang JH, Girona GE, Aramburu IV, Lemke EA. Labeling proteins on live mammalian cells using click chemistry. Nat Protoc 2015; 10:780-91. [DOI: 10.1038/nprot.2015.045] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
200
|
Plasticity of intact rubral projections mediates spontaneous recovery of function after corticospinal tract injury. J Neurosci 2015; 35:1443-57. [PMID: 25632122 DOI: 10.1523/jneurosci.3713-14.2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Axons in the adult CNS fail to regenerate after injury, and therefore recovery from spinal cord injury (SCI) is limited. Although full recovery is rare, a modest degree of spontaneous recovery is observed consistently in a broad range of clinical and nonclinical situations. To define the mechanisms mediating spontaneous recovery of function after incomplete SCI, we created bilaterally complete medullary corticospinal tract lesions in adult mice, eliminating a crucial pathway for voluntary skilled movement. Anatomic and pharmacogenetic tools were used to identify the pathways driving spontaneous functional recovery in wild-type and plasticity-sensitized mice lacking Nogo receptor 1. We found that plasticity-sensitized mice recovered 50% of normal skilled locomotor function within 5 weeks of lesion. This significant, yet incomplete, spontaneous recovery was accompanied by extensive sprouting of intact rubrofugal and rubrospinal projections with the emergence of a de novo circuit between the red nucleus and the nucleus raphe magnus. Transient silencing of this rubro-raphe circuit in vivo via activation of the inhibitory DREADD (designer receptor exclusively activated by designer drugs) receptor hM4di abrogated spontaneous functional recovery. These data highlight the pivotal role of uninjured motor circuit plasticity in supporting functional recovery after trauma, and support a focus of experimental strategies on enhancing intact circuit rearrangement to promote functional recovery after SCI.
Collapse
|