1
|
Chesnokova E, Bal N, Alhalabi G, Balaban P. Regulatory Elements for Gene Therapy of Epilepsy. Cells 2025; 14:236. [PMID: 39937026 PMCID: PMC11816724 DOI: 10.3390/cells14030236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The problem of drug resistance in epilepsy means that in many cases, a surgical treatment may be advised. But this is only possible if there is an epileptic focus, and resective brain surgery may have adverse side effects. One of the promising alternatives is gene therapy, which allows the targeted expression of therapeutic genes in different brain regions, and even in specific cell types. In this review, we provide detailed explanations of some key terms related to genetic engineering, and describe various regulatory elements that have already been used in the development of different approaches to treating epilepsy using viral vectors. We compare a few universal promoters for their strength and duration of transgene expression, and in our description of cell-specific promoters, we focus on elements driving expression in glutamatergic neurons, GABAergic neurons and astrocytes. We also explore enhancers and some other cis-regulatory elements currently used in viral vectors for gene therapy, and consider future perspectives of state-of-the-art technologies for designing new, stronger and more specific regulatory elements. Gene therapy has multiple advantages and should become more common in the future, but there is still a lot to study and invent in this field.
Collapse
Affiliation(s)
- Ekaterina Chesnokova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| | - Natalia Bal
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| | - Ghofran Alhalabi
- Laboratory of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia;
| | - Pavel Balaban
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow 117485, Russia; (E.C.); (P.B.)
| |
Collapse
|
2
|
Taguchi J, Yamada Y, Ohta S, Nakasuka F, Yamamoto T, Ozawa M, Yamada Y. A versatile in vivo platform for reversible control of transgene expression in adult tissues. Stem Cell Reports 2025; 20:102373. [PMID: 39642874 PMCID: PMC11784451 DOI: 10.1016/j.stemcr.2024.11.003] [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] [Received: 07/02/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 12/09/2024] Open
Abstract
Temporal control of transgenes has advanced biomedical interventions, including in vivo reprogramming, often utilizing the doxycycline (Dox)-mediated Tet-ON system. Here, we developed the Dox-mediated Tet-ON or complementary Tet-OFF counterpart to thoroughly investigate spatial and temporal transgene regulation in adult tissues, revealing inherent limitations and unexpected capabilities of each system. In stark contrast with the Tet-ON system, which was effective only in particular tissues and cell types, primarily epithelial cells, the Tet-OFF system proved capable of gene induction across diverse cell types. Despite the drawback of the Tet-OFF system in inducibility and tunability identified in our study, we demonstrated that use of tetracycline (Tc) effectively addresses these issues, possibly through its pharmacologic properties. Our data suggest that the Tc-mediated Tet-OFF system not only enables more versatile control of transgene expression but also offers a more biocompatible alternative for in vivo applications such as tissue regeneration and organismal rejuvenation.
Collapse
Affiliation(s)
- Jumpei Taguchi
- Core Laboratory for Developing Advanced Animal Models, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Yosuke Yamada
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sho Ohta
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Fumie Nakasuka
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Manabu Ozawa
- Core Laboratory for Developing Advanced Animal Models, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Yasuhiro Yamada
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
3
|
Guichardaz M, Bottini S, Balmas E, Bertero A. Overcoming the Silencing of Doxycycline-Inducible Promoters in hiPSC-derived Cardiomyocytes. OPEN RESEARCH EUROPE 2024; 4:266. [PMID: 39926351 PMCID: PMC11803382 DOI: 10.12688/openreseurope.19024.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 02/11/2025]
Abstract
Background Human induced pluripotent stem cells (hiPSCs) are pivotal for studying human development, modeling diseases, and advancing regenerative medicine. Effective control of transgene expression is crucial to achieve temporal and quantitative precision in all of these contexts. The doxycycline (dox)-inducible OPTi-OX system, which integrates the Tet-On 3G transactivator and dox-responsive transgene at the hROSA26 and AAVS1 genomic safe harbors (GSHs), respectively, offers a promising solution. Yet, transgene silencing, particularly in hiPSC-derived cardiomyocytes (hiPSC-CMs), limits its utility. Methods To address this, we evaluated strategies to enhance dox-inducible transgene expression. We compared two promoters, TRE3VG and T11, for activity and stability, and investigated the addition of a Ubiquitous Chromatin Opening Element (UCOE) to reduce silencing. We also tested relocating the transgene cassette to the CLYBL GSH, and employed sodium butyrate (SB), a histone deacetylase inhibitor, to restore promoter activity. Transgene expression was assessed via flow cytometry and real-time quantitative PCR. Results TRE3VG exhibited higher activity than T11, but both were prone to silencing. UCOE did not enhance promoter activity in hiPSCs, but modestly reduced silencing in hiPSC-CMs. Targeting the CLYBL locus improved promoter activity compared to AAVS1 in both hiPSCs and hiPSC-CMs. SB restored activity in silenced inducible promoters within hiPSC-CMs, but compromised hiPSC viability. Unexpectedly, Tet-On 3G was silenced in some clones and could not be reactivated by SB. Conclusions These findings underscore the need for integrating multiple strategies, including careful GSH selection, improved cassette design, epigenetic modulation, and clone screening, to develop robust dox-inducible systems that retain functionality during hiPSC differentiation.
Collapse
Affiliation(s)
- Michelle Guichardaz
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| | - Sveva Bottini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| | - Elisa Balmas
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Torino, 10126, Italy
| |
Collapse
|
4
|
Xu M, Fang M, Chen Q, Xiao W, Xu Z, Cai B, Zhao Z, Wang T, Zhu Z, Chen Y, Zhu Y, Dai M, Jiang T, Li X, Chun S, Zhou R, Li Y, Gou Y, He J, Luo L, You L, Jiang X. GMMID: genetically modified mice information database. Database (Oxford) 2024; 2024:baae078. [PMID: 39163546 PMCID: PMC11334936 DOI: 10.1093/database/baae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 06/19/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Genetically engineered mouse models (GEMMs) are vital for elucidating gene function and disease mechanisms. An overwhelming number of GEMM lines have been generated, but endeavors to collect and organize the information of these GEMMs are seriously lagging behind. Only a few databases are developed for the information of current GEMMs, and these databases lack biological descriptions of allele compositions, which poses a challenge for nonexperts in mouse genetics to interpret the genetic information of these mice. Moreover, these databases usually do not provide information on human diseases related to the GEMM, which hinders the dissemination of the insights the GEMM provides as a human disease model. To address these issues, we developed an algorithm to annotate all the allele compositions that have been reported with Python programming and have developed the genetically modified mice information database (GMMID; http://www.gmmid.cn), a user-friendly database that integrates information on GEMMs and related diseases from various databases, including National Center for Biotechnology Information, Mouse Genome Informatics, Online Mendelian Inheritance in Man, International Mouse Phenotyping Consortium, and Jax lab. GMMID provides comprehensive genetic information on >70 055 alleles, 65 520 allele compositions, and ∼4000 diseases, along with biologically meaningful descriptions of alleles and allele combinations. Furthermore, it provides spatiotemporal visualization of anatomical tissues mentioned in these descriptions, shown alongside the allele compositions. Compared to existing mouse databases, GMMID considers the needs of researchers across different disciplines and presents obscure genetic information in an intuitive and easy-to-understand format. It facilitates users in obtaining complete genetic information more efficiently, making it an essential resource for cross-disciplinary researchers. Database URL: http://www.gmmid.cn.
Collapse
Affiliation(s)
- Menglin Xu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Minghui Fang
- School of Intelligent Systems Engineering, Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
- Guangdong Key Laboratory of Intelligent Transportation Systems (ITS), Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Qiyang Chen
- School of Intelligent Systems Engineering, Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
- Guangdong Key Laboratory of Intelligent Transportation Systems (ITS), Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Wenjun Xiao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Zhixuan Xu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Bao Cai
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Zhenyang Zhao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Tao Wang
- Guangdong GemPharmatech Co,Ltd, No 6, Qianjin West Rd. Shishan Town Nanhai District, Foshan, Guangdong 528225, China
| | - Zhu Zhu
- Guangdong GemPharmatech Co,Ltd, No 6, Qianjin West Rd. Shishan Town Nanhai District, Foshan, Guangdong 528225, China
| | - Yingshan Chen
- Guangdong GemPharmatech Co,Ltd, No 6, Qianjin West Rd. Shishan Town Nanhai District, Foshan, Guangdong 528225, China
| | - Yue Zhu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Mingzhou Dai
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Tiancheng Jiang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Xinyi Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Siuwing Chun
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Runhua Zhou
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Yafei Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Yueyue Gou
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Jingjing He
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Lin Luo
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Linlin You
- School of Intelligent Systems Engineering, Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
- Guangdong Key Laboratory of Intelligent Transportation Systems (ITS), Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| | - Xuan Jiang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, 66#, Gongchang Road, Shenzhen, Guangdong 518107, China
| |
Collapse
|
5
|
Hong J, Sohn KC, Park HW, Jeon H, Ju E, Lee JG, Lee JS, Rho J, Hur GM, Ro H. All-in-one IQ toggle switches with high versatilities for fine-tuning of transgene expression in mammalian cells and tissues. Mol Ther Methods Clin Dev 2024; 32:101202. [PMID: 38374964 PMCID: PMC10875299 DOI: 10.1016/j.omtm.2024.101202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024]
Abstract
The transgene toggling device is recognized as a powerful tool for gene- and cell-based biological research and precision medicine. However, many of these devices often operate in binary mode, exhibit unacceptable leakiness, suffer from transgene silencing, show cytotoxicity, and have low potency. Here, we present a novel transgene switch, SIQ, wherein all the elements for gene toggling are packed into a single vector. SIQ has superior potency in inducing transgene expression in response to tebufenozide compared with the Gal4/UAS system, while completely avoiding transgene leakiness. Additionally, the ease and versatility of SIQ make it possible with a single construct to perform transient transfection, establish stable cell lines by targeting a predetermined genomic locus, and simultaneously produce adenovirus for transduction into cells and mammalian tissues. Furthermore, we integrated a cumate switch into SIQ, called SIQmate, to operate a Boolean AND logic gate, enabling swift toggling-off of the transgene after the removal of chemical inducers, tebufenozide and cumate. Both SIQ and SIQmate offer precise transgene toggling, making them adjustable for various researches, including synthetic biology, genome engineering, and therapeutics.
Collapse
Affiliation(s)
- Jeongkwan Hong
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea (ROK)
| | - Kyung-Cheol Sohn
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 301 747, Korea (ROK)
| | - Hye-Won Park
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea (ROK)
| | - Hyoeun Jeon
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea (ROK)
| | - Eunjin Ju
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 301 747, Korea (ROK)
| | - Jae-Geun Lee
- Microbiome Convergence Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong-Soo Lee
- Microbiome Convergence Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea (ROK)
| | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 301 747, Korea (ROK)
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea (ROK)
| |
Collapse
|
6
|
Pang B, Wu X, Chen H, Yan Y, Du Z, Yu Z, Yang X, Wang W, Lu K. Exploring the memory: existing activity-dependent tools to tag and manipulate engram cells. Front Cell Neurosci 2024; 17:1279032. [PMID: 38259503 PMCID: PMC10800721 DOI: 10.3389/fncel.2023.1279032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/17/2023] [Indexed: 01/24/2024] Open
Abstract
The theory of engrams, proposed several years ago, is highly crucial to understanding the progress of memory. Although it significantly contributes to identifying new treatments for cognitive disorders, it is limited by a lack of technology. Several scientists have attempted to validate this theory but failed. With the increasing availability of activity-dependent tools, several researchers have found traces of engram cells. Activity-dependent tools are based on the mechanisms underlying neuronal activity and use a combination of emerging molecular biological and genetic technology. Scientists have used these tools to tag and manipulate engram neurons and identified numerous internal connections between engram neurons and memory. In this review, we provide the background, principles, and selected examples of applications of existing activity-dependent tools. Using a combination of traditional definitions and concepts of engram cells, we discuss the applications and limitations of these tools and propose certain developmental directions to further explore the functions of engram cells.
Collapse
Affiliation(s)
- Bo Pang
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wu
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Hailun Chen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yiwen Yan
- School of Basic Medicine Science, Southern Medical University, Guangzhou, China
| | - Zibo Du
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zihan Yu
- School of Basic Medicine Science, Southern Medical University, Guangzhou, China
| | - Xiai Yang
- Department of Neurology, Ankang Central Hospital, Ankang, China
| | - Wanshan Wang
- Laboratory Animal Management Center, Southern Medical University, Guangzhou, China
- Guangzhou Southern Medical Laboratory Animal Sci. and Tech. Co., Ltd., Guangzhou, China
| | - Kangrong Lu
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Simunic M, Joshi JT, Merkens H, Colpo N, Kuo HT, Lum JJ, Bénard F. PSMA imaging as a non-invasive tool to monitor inducible gene expression in vivo. EJNMMI Res 2024; 14:3. [PMID: 38177950 PMCID: PMC10767034 DOI: 10.1186/s13550-023-01063-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Affiliation(s)
- Marin Simunic
- Department of Hematology, Clinic for Internal Medicine, Clinical Hospital Centre, Spinciceva 1, 21000, Split, Croatia
| | - Jay T Joshi
- Deeley Research Centre, BC Cancer Research Institute, 2410 Lee Avenue, Victoria, BC, V8R 6V5, Canada
| | - Helen Merkens
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Nadine Colpo
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Hsiou-Ting Kuo
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Julian J Lum
- Deeley Research Centre, BC Cancer Research Institute, 2410 Lee Avenue, Victoria, BC, V8R 6V5, Canada
| | - François Bénard
- BC Cancer Research Institute, 675 West 10Th Avenue, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
8
|
Otomo J, Woltjen K, Sakurai H. Uniform transgene activation in Tet-On systems depends on sustained rtTA expression. iScience 2023; 26:107685. [PMID: 37701566 PMCID: PMC10494183 DOI: 10.1016/j.isci.2023.107685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/13/2023] [Accepted: 08/17/2023] [Indexed: 09/14/2023] Open
Abstract
Application of the tetracycline-inducible gene expression system (Tet-On) in human induced pluripotent stem cells (hiPSCs) has become a fundamental transgenic tool owing to its regulatable gene expression. One of the major hurdles in hiPSC application is non-uniform transgene activation. Here, we report that the supplementation of reverse tetracycline transactivator (rtTA) in polyclonal hiPSCs populations can achieve the uniform transgene activation of Tet-On. Furthermore, the choice of antibiotic selection markers connected by an internal ribosomal entry site (IRES) can influence the expression of upstream transgenes. In particular, expression of the rtTA is more uniform in cell populations when linked to puromycin as compared to neomycin, obviating the need for sub-cloning or supplementation of rtTA. Finally, to expand the range of applications, we adopted our findings to tetracycline-inducible MyoD vector (Tet-MyoD). Our Tet-MyoD promises efficient, robust, and reproducible directed myogenic differentiation of hiPSCs.
Collapse
Affiliation(s)
- Jun Otomo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
9
|
Abdugheni R, Liu C, Liu FL, Zhou N, Jiang CY, Liu Y, Li L, Li WJ, Liu SJ. Comparative genomics reveals extensive intra-species genetic divergence of the prevalent gut commensal Ruminococcus gnavus. Microb Genom 2023; 9:mgen001071. [PMID: 37486746 PMCID: PMC10438805 DOI: 10.1099/mgen.0.001071] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Ruminococcus gnavus is prevalent in the intestines of humans and animals, and ambiguities have been reported regarding its relations with the development of diseases and host well-being. We postulate the ambiguities of its function in different cases may be attributed to strain-level variability of genomic features of R. gnavus. We performed comparative genomic and pathogenicity prediction analysis on 152 filtered high-quality genomes, including 4 genomes of strains isolated from healthy adults in this study. The mean G+C content of genomes of R. gnavus was 42.73±0.33 mol%, and the mean genome size was 3.46±0.34 Mbp. Genome-wide evolutionary analysis revealed R. gnavus genomes were divided into three major phylogenetic clusters. Pan-core genome analysis revealed that there was a total of 28 072 predicted genes, and the core genes, soft-core genes, shell genes and cloud genes accounted for 3.74 % (1051/28 072), 1.75 % (491/28 072), 9.88 % (2774/28 072) and 84.63 % (23 756/28 072) of the total genes, respectively. The small proportion of core genes reflected the wide divergence among R. gnavus strains. We found certain coding sequences with determined health benefits (such as vitamin production and arsenic detoxification), whilst some had an implication of health adversity (such as sulfide dehydrogenase subunits). The functions of the majority of core genes were unknown. The most widespread genes functioning in antibiotic resistance and virulence are tetO (tetracycline-resistance gene, present in 75 strains) and cps4J (capsular polysaccharide biosynthesis protein Cps4J encoding gene, detected in 3 genomes), respectively. Our results revealed genomic divergence and the existence of certain safety-relevant factors of R. gnavus. This study provides new insights for understanding the genomic features and health relevance of R. gnavus, and raises concerns regarding predicted prevalent pathogenicity and antibiotic resistance among most of the strains.
Collapse
Affiliation(s)
- Rashidin Abdugheni
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, PR China
| | - Feng-Lan Liu
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
- College of Life Sciences, Hebei University, Baoding 071000, PR China
| | - Nan Zhou
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Cheng-Ying Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, PR China
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
- University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yonghong Liu
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Li Li
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Wen-Jun Li
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, PR China
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
- University of the Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
10
|
Khayatan D, Hussain A, Tebyaniyan H. Exploring animal models in oral cancer research and clinical intervention: A critical review. Vet Med Sci 2023. [PMID: 37196179 DOI: 10.1002/vms3.1161] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Cancer is a leading cause of death worldwide, but advances in treatment, early detection, and prevention have helped to reduce its impact. To translate cancer research findings into clinical interventions for patients, appropriate animal experimental models, particularly in oral cancer therapy, can be helpful. In vitro experiments using animal or human cells can provide insight into cancer's biochemical pathways. This review discusses the various animal models used in recent years for research and clinical intervention in oral cancer, along with their advantages and disadvantages. We highlight the advantages and limitations of the used animal models in oral cancer research and therapy by searching the terms of animal models, oral cancer, oral cancer therapy, oral cancer research, and animals to find all relevant publications during 2010-2023. Mouse models, widely used in cancer research, can help us understand protein and gene functions in vivo and molecular pathways more deeply. To induce cancer in rodents, xenografts are often used, but companion animals with spontaneous tumours are underutilized for rapid advancement in human and veterinary cancer treatments. Like humans with cancer, companion animals exhibit biological behaviour, treatment responses, and cytotoxic agent responses similar to humans. In companion animal models, disease progression is more rapid, and the animals have a shorter lifespan. Animal models allow researchers to study how immune cells interact with cancer cells and how they can be targeted specifically. Additionally, animal models have been extensively used in research on oral cancers, so researchers can use existing knowledge and tools to better understand oral cancers using animal models.
Collapse
Affiliation(s)
- Danial Khayatan
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Ahmed Hussain
- School of Dentistry, Edmonton Clinic Health Academy (ECHA), University of Alberta, Edmonton, Canada
| | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| |
Collapse
|
11
|
Wei XF, Li S, Hu JL. A TurboID-based proximity labelling approach for identifying the DNA-binding proteins. STAR Protoc 2023; 4:102139. [PMID: 36861822 PMCID: PMC9982669 DOI: 10.1016/j.xpro.2023.102139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 11/04/2022] [Accepted: 02/07/2023] [Indexed: 02/27/2023] Open
Abstract
Biotin proximity labeling is a technique based on the TurboID enzyme that can be used to capture weak or dynamic interactions that had previously not been used to map proteins interacting with a specific DNA sequence. Here, we present a protocol for identifying specific DNA-sequence-binding proteins. We describe steps for biotin labeling of DNA-binding proteins, protein enrichment and sodium dodecyl sulfate polyacrylamide gel electrophoresis separation, and proteomic analysis. For complete details on the use and execution of this protocol, please refer to Wei et al. (2022).1.
Collapse
Affiliation(s)
- Xia-Fei Wei
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China; Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China.
| | - Shan Li
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jie-Li Hu
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Carlton AJ, Jeng J, Grandi FC, De Faveri F, Ceriani F, De Tomasi L, Underhill A, Johnson SL, Legan KP, Kros CJ, Richardson GP, Mustapha M, Marcotti W. A critical period of prehearing spontaneous Ca 2+ spiking is required for hair-bundle maintenance in inner hair cells. EMBO J 2023; 42:e112118. [PMID: 36594367 PMCID: PMC9929643 DOI: 10.15252/embj.2022112118] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 01/04/2023] Open
Abstract
Sensory-independent Ca2+ spiking regulates the development of mammalian sensory systems. In the immature cochlea, inner hair cells (IHCs) fire spontaneous Ca2+ action potentials (APs) that are generated either intrinsically or by intercellular Ca2+ waves in the nonsensory cells. The extent to which either or both of these Ca2+ signalling mechansims are required for IHC maturation is unknown. We find that intrinsic Ca2+ APs in IHCs, but not those elicited by Ca2+ waves, regulate the maturation and maintenance of the stereociliary hair bundles. Using a mouse model in which the potassium channel Kir2.1 is reversibly overexpressed in IHCs (Kir2.1-OE), we find that IHC membrane hyperpolarization prevents IHCs from generating intrinsic Ca2+ APs but not APs induced by Ca2+ waves. Absence of intrinsic Ca2+ APs leads to the loss of mechanoelectrical transduction in IHCs prior to hearing onset due to progressive loss or fusion of stereocilia. RNA-sequencing data show that pathways involved in morphogenesis, actin filament-based processes, and Rho-GTPase signaling are upregulated in Kir2.1-OE mice. By manipulating in vivo expression of Kir2.1 channels, we identify a "critical time period" during which intrinsic Ca2+ APs in IHCs regulate hair-bundle function.
Collapse
Affiliation(s)
| | - Jing‐Yi Jeng
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | | | | | | | | | | | - Stuart L Johnson
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Kevin P Legan
- School of Life SciencesUniversity of Sussex, FalmerBrightonUK
| | - Corné J Kros
- School of Life SciencesUniversity of Sussex, FalmerBrightonUK
| | | | - Mirna Mustapha
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Walter Marcotti
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
| |
Collapse
|
13
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
14
|
Colicchia V, Häggblad M, Sirozh O, Porebski B, Balan M, Li X, Lidemalm L, Carreras‐Puigvert J, Hühn D, Fernandez‐Capetillo O. New regulators of the tetracycline-inducible gene expression system identified by chemical and genetic screens. FEBS Open Bio 2022; 12:1896-1908. [PMID: 36062323 PMCID: PMC9527584 DOI: 10.1002/2211-5463.13482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/01/2022] [Accepted: 09/02/2022] [Indexed: 12/14/2022] Open
Abstract
The tetracycline repressor (tetR)-regulated system is a widely used tool to specifically control gene expression in mammalian cells. Based on this system, we generated a human osteosarcoma cell line, which allows for the inducible expression of an EGFP fusion of the TAR DNA-binding protein 43 (TDP-43), which has been linked to neurodegenerative diseases. Consistent with previous findings, TDP-43 overexpression led to the accumulation of aggregates and limited the viability of U2OS. Using this inducible system, we conducted a chemical screen with a library that included FDA-approved drugs. While the primary screen identified several compounds that prevented TDP-43 toxicity, further experiments revealed that these chemicals abrogated the doxycycline-dependent TDP-43 expression. This antagonistic effect was observed with both doxycycline and tetracycline, and in several Tet-On cell lines expressing different genes, confirming the general effect of these compounds as inhibitors of the tetR system. Using the same cell line, a genome-wide CRISPR/Cas9 screen identified epigenetic regulators such as the G9a methyltransferase and TRIM28 as potential modifiers of TDP-43 toxicity. Yet again, further experiments revealed that G9a inhibition or TRIM28 loss prevented doxycycline-dependent expression of TDP-43. In summary, we have identified new chemical and genetic regulators of the tetR system, thereby raising awareness of the limitations of this approach to conduct chemical or genetic screening in mammalian cells.
Collapse
Affiliation(s)
- Valeria Colicchia
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden,Present address:
Department of BiologyUniversity of Rome Tor VergataItaly
| | - Maria Häggblad
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Oleksandra Sirozh
- Genomic Instability GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Bartlomiej Porebski
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Mirela Balan
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Xuexin Li
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Louise Lidemalm
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Jordi Carreras‐Puigvert
- Present address:
Department of BiologyUniversity of Rome Tor VergataItaly,Present address:
Department of Pharmaceutical Biosciences and Science for Life LaboratoryUppsala UniversitySweden
| | - Daniela Hühn
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Oscar Fernandez‐Capetillo
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden,Genomic Instability GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
15
|
Wei XF, Fan SY, Wang YW, Li S, Long SY, Gan CY, Li J, Sun YX, Guo L, Wang PY, Yang X, Wang JL, Cui J, Zhang WL, Huang AL, Hu JL. Identification of STAU1 as a regulator of HBV replication by TurboID-based proximity labeling. iScience 2022; 25:104416. [PMID: 35663023 PMCID: PMC9156947 DOI: 10.1016/j.isci.2022.104416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 04/21/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
The core promoter (CP) of hepatitis B virus (HBV) is critical for HBV replication by controlling the transcription of pregenomic RNA (pgRNA). Host factors regulating the activity of the CP can be identified by different methods. Biotin-based proximity labeling, a powerful method with the capability to capture weak or dynamic interactions, has not yet been used to map proteins interacting with the CP. Here, we established a strategy, based on the newly evolved promiscuous enzyme TurboID, for interrogating host factors regulating the activity of HBV CP. Using this strategy, we identified STAU1 as an important factor involved in the regulation of HBV CP. Mechanistically, STAU1 indirectly binds to CP mediated by TARDBP, and recruits the SAGA transcription coactivator complex to the CP to upregulate its activity. Moreover, STAU1 binds to HBx and enhances the level of HBx by stabilizing it in a ubiquitin-independent manner.
Collapse
Affiliation(s)
- Xia-Fei Wei
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Shu-Ying Fan
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yu-Wei Wang
- Department of Laboratory Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Shan Li
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shao-Yuan Long
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Chun-Yang Gan
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jie Li
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yu-Xue Sun
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lin Guo
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Pei-Yun Wang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xue Yang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jin-Lan Wang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jing Cui
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wen-Lu Zhang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Jie-Li Hu
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Barykina NV, Karasev MM, Verkhusha VV, Shcherbakova DM. Technologies for large-scale mapping of functional neural circuits active during a user-defined time window. Prog Neurobiol 2022; 216:102290. [PMID: 35654210 DOI: 10.1016/j.pneurobio.2022.102290] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022]
Abstract
The mapping of neural circuits activated during behavior down to individual neurons is crucial for decoding how the brain processes information. Technologies allowing activity-dependent labeling of neurons during user-defined restricted time windows are rapidly developing. Precise marking of the time window with light, in addition to chemicals, is now possible. In these technologies, genetically encoded molecules integrate molecular events resulting from neuronal activity with light/drug-dependent events. The outputs are either changes in fluorescence or activation of gene expression. Molecular reporters allow labeling of activated neurons for visualization and cell-type identification. The transcriptional readout also allows further control of activated neuronal populations using optogenetic tools as reporters. Here we review the design of these technologies and discuss their demonstrated applications to reveal previously unknown connections in the mammalian brain. We also consider the strengths and weaknesses of the current approaches and provide a perspective for the future.
Collapse
Affiliation(s)
- Natalia V Barykina
- P.K. Anokhin Institute of Normal Physiology, Moscow 125315, Russia; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Maksim M Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Vladislav V Verkhusha
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
17
|
Disruption of PAK3 Signaling in Social Interaction Induced cFos Positive Cells Impairs Social Recognition Memory. Cells 2021; 10:cells10113010. [PMID: 34831234 PMCID: PMC8616103 DOI: 10.3390/cells10113010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
P21-activated kinase 3 (PAK3) gene mutations are linked to several neurodevelopmental disorders, but the underlying mechanisms remain unclear. In this study, we used a tetracycline-inducible system to control the expression of a mutant PAK3 (mPAK3) protein in immediate early gene, namely cFos, positive cells to disrupt PAK signaling, specifically in cells activated by social interaction in transgenic mice. We show that the expression of mPAK3-GFP proteins was in cFos-expressing excitatory and inhibitory neurons in various brain regions, such as the cortex and hippocampus, commonly activated during learning and memory. Basal expression of mPAK3-GFP proteins in cFos-positive cells resulted in social recognition memory deficits in the three-chamber social interaction test, without affecting locomotor activity or other forms of memory. The social memory deficit was rescued by doxycycline to halt the mPAK3-GFP transgene expression. In addition, we show that the expression of mPAK3-GFP proteins in a subset of cFos-positive cells, induced by an antecedent short social interaction, termed social pairing, was sufficient to impair social recognition memory. These results indicate that normal PAK signaling in cFos-positive cells activated during social interaction is critical for social memory.
Collapse
|
18
|
Akter F, Simon B, de Boer NL, Redjal N, Wakimoto H, Shah K. Pre-clinical tumor models of primary brain tumors: Challenges and opportunities. Biochim Biophys Acta Rev Cancer 2021; 1875:188458. [PMID: 33148506 PMCID: PMC7856042 DOI: 10.1016/j.bbcan.2020.188458] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 02/09/2023]
Abstract
Primary brain tumors are a heterogeneous group of malignancies that originate in cells of the central nervous system. A variety of models tractable for preclinical studies have been developed to recapitulate human brain tumors, allowing us to understand the underlying pathobiology and explore potential treatments. However, many promising therapeutic strategies identified using preclinical models have shown limited efficacy or failed at the clinical trial stage. The inability to develop therapeutic strategies that significantly improve survival rates in patients highlight the compelling need to revisit the design of currently available animal models and explore the use of new models that allow us to bridge the gap between promising preclinical findings and clinical translation. In this review, we discuss current strategies used to model glioblastoma, the most malignant brain tumor in adults and highlight the shortcomings of specific models that must be circumvented for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Farhana Akter
- Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Brennan Simon
- Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Nadine Leonie de Boer
- Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Navid Redjal
- Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Hiroaki Wakimoto
- Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, United States of America.
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, United States of America.
| |
Collapse
|
19
|
Abstract
The Cre-LoxP technology permits gene ablation in specific cell lineages, at chosen differentiation stages of this lineage and in an inducible manner. It has allowed tremendous advances in our understanding of skeleton biology and related pathophysiological mechanisms, through the generation of loss/gain of function or cell tracing experiments based on the creation of an expanding toolbox of transgenic mice expressing the Cre recombinase in skeletal stem cells, chondrocytes, osteoblasts, or osteoclasts. In this chapter, we provide an overview of the different Cre-LoxP systems and Cre mouse lines used in the bone field, we discuss their advantages, limitations, and we outline best practices to interpret results obtained from the use of Cre mice.
Collapse
Affiliation(s)
- Florent Elefteriou
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Greig Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
20
|
Rommereim LM, Akhade AS, Dutta B, Hutcheon C, Lounsbury NW, Rostomily CC, Savan R, Fraser IDC, Germain RN, Subramanian N. A small sustained increase in NOD1 abundance promotes ligand-independent inflammatory and oncogene transcriptional responses. Sci Signal 2020; 13:13/661/eaba3244. [PMID: 33293463 DOI: 10.1126/scisignal.aba3244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Small, genetically determined differences in transcription [expression quantitative trait loci (eQTLs)] are implicated in complex diseases through unknown molecular mechanisms. Here, we showed that a small, persistent increase in the abundance of the innate pathogen sensor NOD1 precipitated large changes in the transcriptional state of monocytes. A ~1.2- to 1.3-fold increase in NOD1 protein abundance resulting from loss of regulation by the microRNA cluster miR-15b/16 lowered the threshold for ligand-induced activation of the transcription factor NF-κB and the MAPK p38. An additional sustained increase in NOD1 abundance to 1.5-fold over basal amounts bypassed this low ligand concentration requirement, resulting in robust ligand-independent induction of proinflammatory genes and oncogenes. These findings reveal that tight regulation of NOD1 abundance prevents this sensor from exceeding a physiological switching checkpoint that promotes persistent inflammation and oncogene expression. Furthermore, our data provide insight into how a quantitatively small change in protein abundance can produce marked changes in cell state that can serve as the initiator of disease.
Collapse
Affiliation(s)
| | | | - Bhaskar Dutta
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0421, USA
| | | | - Nicolas W Lounsbury
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0421, USA
| | | | - Ram Savan
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Iain D C Fraser
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0421, USA
| | - Ronald N Germain
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0421, USA
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA 98109, USA. .,Department of Immunology, University of Washington, Seattle, WA 98109, USA.,Department of Global Health, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
21
|
Kim H, Selvaraj S, Kiley J, Azzag K, Garay BI, Perlingeiro RCR. Genomic Safe Harbor Expression of PAX7 for the Generation of Engraftable Myogenic Progenitors. Stem Cell Reports 2020; 16:10-19. [PMID: 33275879 PMCID: PMC7815936 DOI: 10.1016/j.stemcr.2020.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
Inducible expression of PAX7 in differentiating pluripotent stem cells (PSCs) allows massively scalable generation of human myogenic progenitors, which upon transplantation into dystrophic muscles give rise to donor-derived myofibers and satellite cells. Therefore, PSC-derived PAX7+ myogenic progenitors represent an attractive therapeutic approach to promote muscle regeneration. Work to date has used lentiviral vectors (LVs) that randomly integrate inducible PAX7 transgenes. Here, we investigated whether equivalent induction of the myogenic program could be achieved by targeting the PAX7 transgene into genomic safe harbor (GSH) sites. Across multiple PSC lines, we find that this approach consistently generates expandable myogenic progenitors in vitro, although scalability of expansion is moderately reduced compared with the LV approach. Importantly, transplantation of GSH-targeted myogenic progenitors produces robust engraftment, comparable with LV counterparts. These findings provide proof of concept for the use of GSH targeting as a potential alternative approach to generate therapeutic PSC-derived myogenic progenitors for clinical applications.
Collapse
Affiliation(s)
- Hyunkee Kim
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA; Department of Genetic, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA; Department of Genetic, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA
| | - Bayardo I Garay
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 4-128 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA; Department of Genetic, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
22
|
Testing the CRISPR-Cas9 and glmS ribozyme systems in Leishmania tarentolae. Mol Biochem Parasitol 2020; 241:111336. [PMID: 33166572 DOI: 10.1016/j.molbiopara.2020.111336] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 02/03/2023]
Abstract
Leishmania parasites include important pathogens and model organisms and are even used for the production of recombinant proteins. However, functional genomics and the characterization of essential genes are often limited in Leishmania because of low-throughput technologies for gene disruption or tagging and the absence of components for RNA interference. Here, we tested the T7 RNA polymerase-dependent CRISPR-Cas9 system by Beneke et al. and the glmS ribozyme-based knock-down system in the model parasite Leishmania tarentolae. We successfully deleted two reference genes encoding the flagellar motility factor Pf16 and the salvage-pathway enzyme adenine phosphoribosyltransferase, resulting in immotile and drug-resistant parasites, respectively. In contrast, we were unable to disrupt the gene encoding the mitochondrial flavoprotein Erv. Cultivation of L. tarentolae in standard BHI medium resulted in a constitutive down-regulation of an episomal mCherry-glmS reporter by 40 to 60%. For inducible knock-downs, we evaluated the growth of L. tarentolae in alternative media and identified supplemented MEM, IMDM and McCoy's 5A medium as candidates. Cultivation in supplemented MEM allowed an inducible, glucosamine concentration-dependent down-regulation of the episomal mCherry-glmS reporter by more than 70%. However, chromosomal glmS-tagging of the genes encoding Pf16, adenine phosphoribosyltransferase or Erv did not reveal a knock-down phenotype. Our data demonstrate the suitability of the CRISPR-Cas9 system for the disruption and tagging of genes in L. tarentolae as well as the limitations of the glmS system, which was restricted to moderate efficiencies for episomal knock-downs and caused no detectable phenotype for chromosomal knock-downs.
Collapse
|
23
|
Martins NMC, Cisneros-Soberanis F, Pesenti E, Kochanova NY, Shang WH, Hori T, Nagase T, Kimura H, Larionov V, Masumoto H, Fukagawa T, Earnshaw WC. H3K9me3 maintenance on a human artificial chromosome is required for segregation but not centromere epigenetic memory. J Cell Sci 2020; 133:jcs242610. [PMID: 32576667 PMCID: PMC7390644 DOI: 10.1242/jcs.242610] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
Most eukaryotic centromeres are located within heterochromatic regions. Paradoxically, heterochromatin can also antagonize de novo centromere formation, and some centromeres lack it altogether. In order to investigate the importance of heterochromatin at centromeres, we used epigenetic engineering of a synthetic alphoidtetO human artificial chromosome (HAC), to which chimeric proteins can be targeted. By tethering the JMJD2D demethylase (also known as KDM4D), we removed heterochromatin mark H3K9me3 (histone 3 lysine 9 trimethylation) specifically from the HAC centromere. This caused no short-term defects, but long-term tethering reduced HAC centromere protein levels and triggered HAC mis-segregation. However, centromeric CENP-A was maintained at a reduced level. Furthermore, HAC centromere function was compatible with an alternative low-H3K9me3, high-H3K27me3 chromatin signature, as long as residual levels of H3K9me3 remained. When JMJD2D was released from the HAC, H3K9me3 levels recovered over several days back to initial levels along with CENP-A and CENP-C centromere levels, and mitotic segregation fidelity. Our results suggest that a minimal level of heterochromatin is required to stabilize mitotic centromere function but not for maintaining centromere epigenetic memory, and that a homeostatic pathway maintains heterochromatin at centromeres.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
| | | | - Elisa Pesenti
- Wellcome Trust Centre for Cell Biology, Edinburgh, UK
| | | | - Wei-Hao Shang
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Tetsuya Hori
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | | - Hiroshi Kimura
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Vladimir Larionov
- National Cancer Institute, National Institutes of Health, Bethesda, USA
| | | | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | |
Collapse
|
24
|
Systematic microsatellite repeat expansion cloning and validation. Hum Genet 2020; 139:1233-1246. [PMID: 32277284 DOI: 10.1007/s00439-020-02165-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/04/2020] [Indexed: 10/24/2022]
Abstract
Approximately 3% of the human genome is composed of short tandem repeat (STR) DNA sequence known as microsatellites, which can be found in both coding and non-coding regions. When associated with genic regions, expansion of microsatellite repeats beyond a critical threshold causes dozens of neurological repeat expansion disorders. To better understand the molecular pathology of repeat expansion disorders, precise cloning of microsatellite repeat sequence and expansion size is highly valuable. Unfortunately, cloning repeat expansions is often challenging and presents a significant bottleneck to practical investigation. Here, we describe a clear method for seamless and systematic cloning of practically any microsatellite repeat expansion. We use cloning and expansion of GGGGCC repeats, which are the leading genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), as an example. We employ a recursive directional ligation (RDL) technique to build multiple GGGGCC repeat-containing vectors. We describe methods to validate repeat expansion cloning, including diagnostic restriction digestion, PCR across the repeat, and next-generation long-read MinION nanopore sequencing. Validated cloning of microsatellite repeats beyond the critical expansion threshold can facilitate step-by-step characterization of disease mechanisms at the cellular and molecular level.
Collapse
|
25
|
Mori Y, Yoshida Y, Satoh A, Moriya H. Development of an experimental method of systematically estimating protein expression limits in HEK293 cells. Sci Rep 2020; 10:4798. [PMID: 32179769 PMCID: PMC7075890 DOI: 10.1038/s41598-020-61646-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/28/2020] [Indexed: 11/25/2022] Open
Abstract
Protein overexpression sometimes causes cellular defects, although the underlying mechanism is still unknown. A protein's expression limit, which triggers cellular defects, is a useful indication of the underlying mechanism. In this study, we developed an experimental method of estimating the expression limits of target proteins in the human embryonic kidney cell line HEK293 by measuring the proteins' expression levels in cells that survived after the high-copy introduction of plasmid DNA by which the proteins were expressed under a strong cytomegalovirus promoter. The expression limits of nonfluorescent target proteins were indirectly estimated by measuring the levels of green fluorescent protein (GFP) connected to the target proteins with the self-cleaving sequence P2A. The expression limit of a model GFP was ~5.0% of the total protein, and sustained GFP overexpression caused cell death. The expression limits of GFPs with mitochondria-targeting signals and endoplasmic reticulum localization signals were 1.6% and 0.38%, respectively. The expression limits of four proteins involved in vesicular trafficking were far lower compared to a red fluorescent protein. The protein expression limit estimation method developed will be valuable for defining toxic proteins and consequences of protein overexpression.
Collapse
Affiliation(s)
- Yoshihiro Mori
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Yuki Yoshida
- Sony Computer Science Laboratories, Tokyo, Japan
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Hisao Moriya
- Research Core for Interdisciplinary Sciences, Okayama University, Okayama, Japan.
- Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan.
| |
Collapse
|
26
|
Tsujimura M, Kusamori K, Nishikawa M. Rapid Regulation of Human Mesenchymal Stem Cell Proliferation Using Inducible Caspase-9 Suicide Gene for Safe Cell-Based Therapy. Int J Mol Sci 2019; 20:ijms20225759. [PMID: 31744061 PMCID: PMC6887989 DOI: 10.3390/ijms20225759] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/01/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022] Open
Abstract
The regulation of transplanted cell proliferation and function is important to achieve safe cell-based therapies. We previously reported that the proliferation and function of transplanted cells, which expressed the herpes simplex virus thymidine kinase (HSVtk) suicide gene, could be controlled by ganciclovir (GCV) administration. However, there are some concerns regarding the use of GCV. It is reported that the inducible caspase-9 (iC9) gene, a human caspase-9-derived genetically engineered suicide gene, rapidly induces cell apoptosis in the presence of apoptosis inducers, such as AP20187. In this study, we used a combination of the iC9 gene and AP20187 to achieve rapid regulation of transplanted cell proliferation. Cells from the human mesenchymal stem cell line UE7T-13 were transfected with the iC9 gene to obtain UE7T-13/iC9 cells. AP20187 significantly reduced the number of UE7T-13/iC9 cells within 24 h in a concentration-dependent manner. This reduction was much faster than the reduction of HSVtk-expressing UE7T-13 cells induced by GCV addition. Subcutaneous AP20187 administration rapidly reduced the luminescence signal from NanoLuc luciferase (Nluc)-expressing UE7T-13/iC9 cells transplanted into mice. These results indicate that the combined use of the iC9 gene and AP20187 is effective in rapidly regulating transplanted cell proliferation.
Collapse
Affiliation(s)
| | - Kosuke Kusamori
- Correspondence: ; Tel.: +81-4-7124-1501; Fax: +81-4-7121-4450
| | | |
Collapse
|
27
|
Monitoring method for transgene expression in target tissue by blood sampling. ACTA ACUST UNITED AC 2019; 24:e00401. [PMID: 31788440 PMCID: PMC6880019 DOI: 10.1016/j.btre.2019.e00401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 01/06/2023]
Abstract
Transgene expressions of simultaneously-administered two plasmid DNA in muscle correlated each other. Transgene expressions of secretable luciferase in muscle and plasma also correlated each other. It was possible to monitor transgene expression in tissues by blood sampling.
In this study, we have developed a novel method to monitor transgene expression in tissues by blood sampling. We administered plasmid DNA (pDNA) encoding non-secretory form of firefly luciferase as a reporter gene and pDNA encoding secretable Gaussia princeps luciferase as a monitor gene simultaneously into mice. Good positive correlations were found between log-transgene expression of the reporter gene and the monitor gene in the treated muscle, between the monitor gene in the treated muscle and plasma, and consequently between the reporter gene in the treated muscle and the monitor gene in plasma after naked pDNA transfer into the muscle of mice. Such positive correlations were also found with gastric serosal surface instillation of naked pDNA, intravenous injection of lipoplex, and hydrodynamics-based injection of naked pDNA. We developed monitoring method of transgene expression in tissues by blood sampling, which was named ‘Therapeutic transgene monitoring (TTM)’, after ‘Therapeutic drug monitoring (TDM)’.
Collapse
|
28
|
Pedone E, Postiglione L, Aulicino F, Rocca DL, Montes-Olivas S, Khazim M, di Bernardo D, Pia Cosma M, Marucci L. A tunable dual-input system for on-demand dynamic gene expression regulation. Nat Commun 2019; 10:4481. [PMID: 31578371 PMCID: PMC6775159 DOI: 10.1038/s41467-019-12329-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular systems have evolved numerous mechanisms to adapt to environmental stimuli, underpinned by dynamic patterns of gene expression. In addition to gene transcription regulation, modulation of protein levels, dynamics and localization are essential checkpoints governing cell functions. The introduction of inducible promoters has allowed gene expression control using orthogonal molecules, facilitating its rapid and reversible manipulation to study gene function. However, differing protein stabilities hinder the generation of protein temporal profiles seen in vivo. Here, we improve the Tet-On system integrating conditional destabilising elements at the post-translational level and permitting simultaneous control of gene expression and protein stability. We show, in mammalian cells, that adding protein stability control allows faster response times, fully tunable and enhanced dynamic range, and improved in silico feedback control of gene expression. Finally, we highlight the effectiveness of our dual-input system to modulate levels of signalling pathway components in mouse Embryonic Stem Cells.
Collapse
Affiliation(s)
- Elisa Pedone
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK.
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| | - Lorena Postiglione
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Francesco Aulicino
- BrisSynBio, Bristol, BS8 1TQ, UK
- Department of Biochemistry, Bristol, BS8 1TD, UK
| | - Dan L Rocca
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK.
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
- BrisSynBio, Bristol, BS8 1TQ, UK.
| | - Sandra Montes-Olivas
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK
| | - Mahmoud Khazim
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine Via Campi Flegrei 34, 80078, Pozzuoli, Italy
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), Dr Aiguader 88, 08002, Barcelona, Spain
- Universitati Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Luis Companys, 08010, Barcelona, Spain
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), 510005, Guangzhou, China
- Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, 510530, Guangzhou, China
| | - Lucia Marucci
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK.
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
- BrisSynBio, Bristol, BS8 1TQ, UK.
| |
Collapse
|
29
|
Davies JA, Ireland S, Harding S, Sharman JL, Southan C, Dominguez-Monedero A. Inverse pharmacology: Approaches and tools for introducing druggability into engineered proteins. Biotechnol Adv 2019; 37:107439. [PMID: 31494210 PMCID: PMC6891246 DOI: 10.1016/j.biotechadv.2019.107439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/24/2019] [Accepted: 08/20/2019] [Indexed: 01/08/2023]
Abstract
A major feature of twenty-first century medical research is the development of therapeutic strategies that use 'biologics' (large molecules, usually engineered proteins) and living cells instead of, or as well as, the small molecules that were the basis of pharmacology in earlier eras. The high power of these techniques can bring correspondingly high risk, and therefore the need for the potential for external control. One way of exerting control on therapeutic proteins is to make them responsive to small molecules; in a clinical context, these small molecules themselves have to be safe. Conventional pharmacology has resulted in thousands of small molecules licensed for use in humans, and detailed structural data on their binding to their protein targets. In principle, these data can be used to facilitate the engineering of drug-responsive modules, taken from natural proteins, into synthetic proteins. This has been done for some years (for example, Cre-ERT2) but usually in a painstaking manner. Recently, we have developed the bioinformatic tool SynPharm to facilitate the design of drug-responsive proteins. In this review, we outline the history of the field, the design and use of the Synpharm tool, and describe our own experiences in engineering druggability into the Cpf1 effector of CRISPR gene editing.
Collapse
Affiliation(s)
- Jamie A Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XB, UK.
| | - Sam Ireland
- Biomolecular Structure & Modelling Unit, Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Simon Harding
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XB, UK
| | - Joanna L Sharman
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XB, UK; Novo Nordisk Research Centre Oxford, Novo Nordisk Ltd, Innovation Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7FZ, UK
| | | | - Alazne Dominguez-Monedero
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XB, UK
| |
Collapse
|
30
|
Wang Y, Zhang K, Li H, Xu X, Xue H, Wang P, Fu YV. Fine-tuning the expression of target genes using a DDI2 promoter gene switch in budding yeast. Sci Rep 2019; 9:12538. [PMID: 31467340 PMCID: PMC6715627 DOI: 10.1038/s41598-019-49000-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/21/2019] [Indexed: 11/22/2022] Open
Abstract
Tuned gene expression is crucial to the proper growth and response to the environmental changes of an organism. To enable tunable gene expression as designed is desirable in both scientific research and industrial application. Here, we introduce a novel promoter switching method based on the DDI2 promoter (PDDI2) that can fine tune the expression of target genes. We constructed a recyclable cassette (PDDI2-URA3-PDDI2) and integrated it upstream of yeast target genes to replace the native promoters by DDI2 promoter without introducing any junk sequence. We found that the presence or absence of cyanamide as an inducer could turn on or off the expression of target genes. In addition, we showed that PDDI2 could act as a gene switch to linearly regulate the expression levels of target genes in vivo. We switched the original promoters of RAD18, TUP1, and CDC6 with PDDI2 as a proof-of-concept.
Collapse
Affiliation(s)
- Yong Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Kaining Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Hanfei Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xin Xu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Huijun Xue
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pingping Wang
- Qingdao Baihuizhiye Biotech Co.Ltd, Qingdao, 266109, China
| | - Yu V Fu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China. .,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
31
|
Evidence that Ergosterol Biosynthesis Modulates Activity of the Pdr1 Transcription Factor in Candida glabrata. mBio 2019; 10:mBio.00934-19. [PMID: 31186322 PMCID: PMC6561024 DOI: 10.1128/mbio.00934-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A likely contributor to the increased incidence of non-albicans candidemias involving Candida glabrata is the ease with which this yeast acquires azole resistance, in large part due to induction of the ATP-binding cassette transporter-encoding gene CDR1. Azole drugs lead to induction of Pdr1 transactivation, with a central model being that this factor binds these drugs directly. Here we provide evidence that Pdr1 is activated without azole drugs by the use of genetic means to inhibit expression of azole drug target-encoding gene ERG11. These acute reductions in Erg11 levels lead to elevated Pdr1 activity even though no drug is present. A key transcriptional regulator of the ERG pathway, Upc2A, is shown to directly bind to the PDR1 and CDR1 promoters. We interpret these data as support for the view that Pdr1 function is responsive to ergosterol biosynthesis and suggest that this connection reveals the normal physiological circuitry in which Pdr1 participates. A crucial limitation in antifungal chemotherapy is the limited number of antifungal drugs currently available. Azole drugs represent the most commonly used chemotherapeutic, and loss of efficacy of these drugs is a major risk factor in successful treatment of a variety of fungal diseases. Candida glabrata is a pathogenic yeast that is increasingly found associated with bloodstream infections, a finding likely contributed to by its proclivity to develop azole drug resistance. C. glabrata often acquires azole resistance via gain-of-function (GOF) mutations in the transcription factor Pdr1. These GOF forms of Pdr1 drive elevated expression of target genes, including the ATP-binding cassette transporter-encoding CDR1 locus. GOF alleles of PDR1 have been extensively studied, but little is known of how Pdr1 is normally regulated. Here we test the idea that reduction of ergosterol biosynthesis (as occurs in the presence of azole drugs) might trigger activation of Pdr1 function. Using two different means of genetically inhibiting ergosterol biosynthesis, we demonstrated that Pdr1 activity and target gene expression are elevated in the absence of azole drug. Blocks at different points in the ergosterol pathway lead to Pdr1 activation as well as to induction of other genes in this pathway. Delivery of the signal from the ergosterol pathway to Pdr1 involves the transcription factor Upc2A, an ERG gene regulator. We show that Upc2A binds directly to the PDR1 and CDR1 promoters. Our studies argue for a physiological link between ergosterol biosynthesis and Pdr1-dependent gene regulation that is not restricted to efflux of azole drugs.
Collapse
|
32
|
Polverini PJ, Lingen MW. A History of Innovations in the Diagnosis and Treatment of Oral and Head and Neck Cancer. J Dent Res 2019; 98:489-497. [PMID: 31008698 DOI: 10.1177/0022034519833645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Historical records as far back as 3000 BCE show that oral and head and neck cancer was a disease process well known to Egyptian physicians. Luminaries such as Hippocrates, Galen, Pott, and Virchow were instrumental in shaping our understanding of the etiology and pathogenesis of cancer. During the 20th century, evidence-based medicine catalyzed the development of rigorous science-based diagnostic and treatment protocols. The use of surgery, therapeutic radiation, and chemotherapy as single-treatment agents or in combination with one another gradually emerged as the preferred approach to cancer therapy. The recognition of tobacco, alcohol, and human papillomavirus as etiological agents in oral and head and neck cancer prompted the development of new diagnostic aids and treatment strategies to mitigate cancer progression. More in-depth mechanistic insights into the multistep process of oral and head and neck cancer were made possible by the use of the hamster buccal pouch and mouse models. New technologies, such as the sequencing of the human genome, metabolomics, and proteomics, have provided the foundation for what we today call precision medicine. The future success of tailored medical treatment for cancer patients will depend on the discovery of new druggable targets with improved therapeutic efficacy. As the precision and sensitivity of existing tools for prevention and risk assessment improve, greater accuracy will be achieved in predicting health outcomes.
Collapse
Affiliation(s)
- P J Polverini
- 1 Department of Periodontics and Oral Medicine, Division of Oral Medicine, Pathology, and Radiology, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,2 Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,3 University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - M W Lingen
- 4 Department of Pathology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
33
|
Couasnay G, Frey C, Elefteriou F. Promoter Cre-Specific Genotyping Assays for Authentication of Cre-Driver Mouse Lines. JBMR Plus 2019; 3:e10128. [PMID: 31044186 PMCID: PMC6478581 DOI: 10.1002/jbm4.10128] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/17/2018] [Accepted: 10/21/2018] [Indexed: 11/07/2022] Open
Abstract
The Cre-LoxP system gene knockout (KO) technology provides cell- and time-specificity of gene ablation to investigate cell-autonomous gene function in vivo, and is paramount for understanding the function of genes involved in bone development, remodeling, and repair. This approach permits gene ablation in a cell- or tissue-specific, differentiation stage-specific, and inducible manner, thanks to the use of well-chosen promoters that drive expression of the Cre recombinase in selected cells/tissues. The generation of these powerful tools has led to the expansion of Cre mouse lines available to the research community, which are often shared within and between laboratories. Although convenient and commonly used, genotyping these Cre lines with a generic set of primers that amplifies the Cre transgene does not distinguish between various Cre-deleter lines. This practice poses the significant risk of mistakenly swapping Cre lineages, as laboratories often host and handle several lines at a time and utilize multiple lines per project. In line with the NIH-led effort to promote authentication of biological reagents and increase scientific rigor, we report here strategies for designing appropriate sets of primers able to discriminate some of most widely used Cre-deleter mouse lines in the field of bone biology, and the validation of 24 of them.
Collapse
Affiliation(s)
| | | | - Florent Elefteriou
- Department of Orthopedic Surgery
- Departments of Human and Molecular GeneticsBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
34
|
Gardinier JD, Daly-Seiler C, Rostami N, Kundal S, Zhang C. Loss of the PTH/PTHrP receptor along the osteoblast lineage limits the anabolic response to exercise. PLoS One 2019; 14:e0211076. [PMID: 30682096 PMCID: PMC6347174 DOI: 10.1371/journal.pone.0211076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Exercise and physical activity are critical to maintain bone mass and strength throughout life. Both exercise and physical activity subject bone to a unique combination of stimuli in the forms of dynamic loading and a systemic increase in parathyroid hormone (PTH). Although dynamic loading is considered to be the primary osteogenic stimuli, the influence of increasing PTH levels remains unclear. We hypothesize that activation of the PTH/PTH-related peptide type 1 receptor (PPR) along the osteoblast lineage facilitates bone formation and improved mechanical properties in response to exercise. To test this hypothesis, conditional PPR-knockout mice (PPRcKO) were generated in which PPR expression was deleted along the osteoblast lineage under the osterix promoter. At 8-weeks of age, both PPRfl/fl and PPRcKO mice were subjected to treadmill running or sedentary conditions for 5-weeks. Under sedentary conditions, PPRcKO mice displayed significantly less bone mass as well as smaller structural-level strength (yield-load and ultimate load), while tissue level properties were largely unaffected. However, PPRcKO mice exposed to exercise displayed significantly less structural-level and tissue-level mechanical properties when compared to exercised PPRfl/fl mice. Overall, these data demonstrate that PPR expression along the osteoblast lineage is essential for exercise to improve the mechanical properties of cortical bone. Furthermore, the influence of PPR activation on material properties is unique to exercise and not during normal growth and development.
Collapse
Affiliation(s)
- Joseph D. Gardinier
- Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States of America
- * E-mail:
| | - Conor Daly-Seiler
- Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States of America
| | - Niloufar Rostami
- Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States of America
| | - Siddharth Kundal
- Biomedical Physics Department, Wayne State University, Detroit, MI, United States of America
| | - Chunbin Zhang
- Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States of America
| |
Collapse
|
35
|
Lin A, Zeng C, Wang Q, Zhang W, Li M, Hanna M, Xiao W. Utilization of a Strongly Inducible DDI2 Promoter to Control Gene Expression in Saccharomyces cerevisiae. Front Microbiol 2018; 9:2736. [PMID: 30505295 PMCID: PMC6250804 DOI: 10.3389/fmicb.2018.02736] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/25/2018] [Indexed: 11/15/2022] Open
Abstract
Regulating target gene expression is a common method in yeast research. In Saccharomyces cerevisiae, there are several widely used regulated expression systems, such as the GAL and Tet-off systems. However, all current expression systems possess some intrinsic deficiencies. We have previously reported that the DDI2 gene can be induced to very high levels upon cyanamide or methyl methanesulfonate treatment. Here we report the construction of gene expression systems based on the DDI2 promoter in both single- and multi-copy plasmids. Using GFP as a reporter gene, it was demonstrated that the target gene expression could be increased by up to 2,000-fold at the transcriptional level by utilizing the above systems. In addition, a DDI2-based construct was created for promoter shuffling in the budding yeast genome to control endogenous gene expression. Overall, this study offers a set of convenient and highly efficient experimental tools to control target gene expression in budding yeast.
Collapse
Affiliation(s)
- Aiyang Lin
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chuanwen Zeng
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
| | - Qian Wang
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
| | - Wenqing Zhang
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
| | - Mengyao Li
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Michelle Hanna
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Wei Xiao
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
36
|
Lee S, Won M, Hwang RH, Hur GM, Ro H. An Ecdysone Receptor-based Singular Gene Switch for Deliberate Expression of Transgene with Robustness, Reversibility, and Negligible Leakiness. J Vis Exp 2018. [PMID: 29781995 DOI: 10.3791/57494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Precise control of transgene expression is desirable in biological and clinical studies. However, because the binary feature of currently employed gene switches requires the transfer of two therapeutic expression units concurrently into a single cell, the practical application of the system for gene therapy is limited. To simplify the transgene expression system, we generated a gene switch designated as pEUI(+) encompassing a complete set of transgene expression modules in a single vector. Comprising of the GAL4 DNA-binding domain and modified EcR (GvEcR), a minimal VP16 activation domain fused with a GAL4 DNA-binding domain, as well as a modified Drosophila ecdysone receptor (EcR), the newly developed singular gene switch is highly responsive to the administration of a chemical inducer in a time- and dosage-dependent manner. The pEUI(+) vector is a potentially powerful tool for improving the control of transgene expression in both biological research and pre-clinical studies. Here, we present a detailed protocol for modulation of a transient and stable transgene expression using pEUI(+) vector by the treatment of tebufenozide (Teb). Additionally, we share important guidelines for the use of Teb as a chemical inducer.
Collapse
Affiliation(s)
- Seoghyun Lee
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University
| | - Minho Won
- Department of Pharmacology, College of Medicine, Chungnam National University
| | | | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University;
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University;
| |
Collapse
|
37
|
Luo JJ, Young CD, Zhou HM, Wang XJ. Mouse Models for Studying Oral Cancer: Impact in the Era of Cancer Immunotherapy. J Dent Res 2018; 97:683-690. [PMID: 29649368 DOI: 10.1177/0022034518767635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Model systems for oral cancer research have progressed from tumor epithelial cell cultures to in vivo systems that mimic oral cancer genetics, pathological characteristics, and tumor-stroma interactions of oral cancer patients. In the era of cancer immunotherapy, it is imperative to use model systems to test oral cancer prevention and therapeutic interventions in the presence of an immune system and to discover mechanisms of stromal contributions to oral cancer carcinogenesis. Here, we review in vivo mouse model systems commonly used for studying oral cancer and discuss the impact these models are having in advancing basic mechanisms, chemoprevention, and therapeutic intervention of oral cancer while highlighting recent discoveries concerning the role of immune cells in oral cancer. Improvements to in vivo model systems that highly recapitulate human oral cancer hold the key to identifying features of oral cancer initiation, progression, and invasion as well as molecular and cellular targets for prevention, therapeutic response, and immunotherapy development.
Collapse
Affiliation(s)
- J J Luo
- 1 State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.,2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - C D Young
- 2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - H M Zhou
- 1 State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - X J Wang
- 2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
38
|
Ambri F, Snoek T, Skjoedt ML, Jensen MK, Keasling JD. Design, Engineering, and Characterization of Prokaryotic Ligand-Binding Transcriptional Activators as Biosensors in Yeast. Methods Mol Biol 2018; 1671:269-290. [PMID: 29170965 DOI: 10.1007/978-1-4939-7295-1_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In cell factory development, screening procedures, often relying on low-throughput analytical methods, are lagging far behind diversity generation methods. This renders the identification and selection of the best cell factory designs tiresome and costly, conclusively hindering the manufacturing process. In the yeast Saccharomyces cerevisiae, implementation of allosterically regulated transcription factors from prokaryotes as metabolite biosensors has proven a valuable strategy to alleviate this screening bottleneck. Here, we present a protocol to select and incorporate prokaryotic transcriptional activators as metabolite biosensors in S. cerevisiae. As an example, we outline the engineering and characterization of the LysR-type transcriptional regulator (LTTR) family member BenM from Acetinobacter sp. ADP1 for monitoring accumulation of cis,cis-muconic acid, a bioplast precursor, in yeast by means of flow cytometry.
Collapse
Affiliation(s)
- Francesca Ambri
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Tim Snoek
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mette L Skjoedt
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Michael K Jensen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Jay D Keasling
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.,Joint BioEnergy Institute, Emeryville, CA, USA.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA.,Department of Bioengineering, University of California, Berkeley, CA, USA
| |
Collapse
|
39
|
Boynton FDD, Ericsson AC, Uchihashi M, Dunbar ML, Wilkinson JE. Doxycycline induces dysbiosis in female C57BL/6NCrl mice. BMC Res Notes 2017; 10:644. [PMID: 29187243 PMCID: PMC5708113 DOI: 10.1186/s13104-017-2960-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE This study aims to demonstrate the effect of oral doxycycline on fecal microbiota of mice. Doxycycline is a common effector for control of gene expression using the tet-inducible system in transgenic mice. The effect of oral doxycycline on murine gut microbiota has not been reported. We evaluated the effect of doxycycline treatment by sequencing the V4 hypervariable region of the 16S rRNA gene from fecal samples collected during a 4 week course of treatment at a dose of 2 mg/ml in the drinking water. RESULTS The fecal microbiota of treated animals were distinct from control animals; the decreased richness and diversity were characterized primarily by Bacteroides sp. enrichment. These effects persisted when the treatment was temporarily discontinued for 1 week. These data suggest that doxycycline treatment can induce significant dysbiosis, and its effects should be considered when used in animal models that are or maybe sensitive to perturbation of the gut microbiota.
Collapse
Affiliation(s)
- Felicia D. Duke Boynton
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
- Research Animal Resources, University of Minnesota, Minneapolis, MN USA
| | - Aaron C. Ericsson
- University of Missouri Metagenomics Research Center, University of Missouri, Columbia, MO USA
| | - Mayu Uchihashi
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
- Medtronic Innovation Center Japan, Medtronic Japan Co., Ltd. Kawasaki, Kanagawa, Japan
| | - Misha L. Dunbar
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
- Research Animal Resources, University of Minnesota, Minneapolis, MN USA
| | - J. Erby Wilkinson
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI USA
| |
Collapse
|
40
|
Das AT, Tenenbaum L, Berkhout B. Tet-On Systems For Doxycycline-inducible Gene Expression. Curr Gene Ther 2017; 16:156-67. [PMID: 27216914 PMCID: PMC5070417 DOI: 10.2174/1566523216666160524144041] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/03/2016] [Accepted: 05/03/2016] [Indexed: 11/22/2022]
Abstract
The tetracycline-controlled Tet-Off and Tet-On gene expression systems are used to regulate the activity of genes in eukaryotic cells in diverse settings, varying from basic biological research to biotechnology and gene therapy applications. These systems are based on regulatory elements that control the activity of the tetracycline-resistance operon in bacteria. The Tet-Off system allows silencing of gene expression by administration of tetracycline (Tc) or tetracycline-derivatives like doxycycline (dox), whereas the Tet-On system allows activation of gene expression by dox. Since the initial design and construction of the original Tet-system, these bacterium-derived systems have been significantly improved for their function in eukaryotic cells. We here review how a dox-controlled HIV-1 variant was designed and used to greatly improve the activity and dox-sensitivity of the rtTA transcriptional activator component of the Tet-On system. These optimized rtTA variants require less dox for activation, which will reduce side effects and allow gene control in tissues where a relatively low dox level can be reached, such as the brain.
Collapse
Affiliation(s)
- Atze T Das
- Laboratory of Experimental Virology, Academic Medical Center, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | | | | |
Collapse
|
41
|
Reid CA, Boye SL, Hauswirth WW, Lipinski DM. miRNA-mediated post-transcriptional silencing of transgenes leads to increased adeno-associated viral vector yield and targeting specificity. Gene Ther 2017; 24:462-469. [DOI: 10.1038/gt.2017.50] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/05/2017] [Accepted: 06/08/2017] [Indexed: 12/21/2022]
|
42
|
Inducible and Deterministic Forward Programming of Human Pluripotent Stem Cells into Neurons, Skeletal Myocytes, and Oligodendrocytes. Stem Cell Reports 2017; 8:803-812. [PMID: 28344001 PMCID: PMC5390118 DOI: 10.1016/j.stemcr.2017.02.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 02/17/2017] [Accepted: 02/17/2017] [Indexed: 12/31/2022] Open
Abstract
The isolation or in vitro derivation of many human cell types remains challenging and inefficient. Direct conversion of human pluripotent stem cells (hPSCs) by forced expression of transcription factors provides a potential alternative. However, deficient inducible gene expression in hPSCs has compromised efficiencies of forward programming approaches. We have systematically optimized inducible gene expression in hPSCs using a dual genomic safe harbor gene-targeting strategy. This approach provides a powerful platform for the generation of human cell types by forward programming. We report robust and deterministic reprogramming of hPSCs into neurons and functional skeletal myocytes. Finally, we present a forward programming strategy for rapid and highly efficient generation of human oligodendrocytes. Dual genomic safe harbor targeting of the Tet-ON system Optimized inducible transgene expression in human pluripotent stem cells Deterministic forward programming into neurons, myocytes, and oligodendrocytes
Collapse
|
43
|
Wang N, Yuan Z, Niu W, Li Q, Guo J. Synthetic biology approach for the development of conditionally replicating HIV-1 vaccine. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2017; 92:455-462. [PMID: 28983143 PMCID: PMC5624719 DOI: 10.1002/jctb.5174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
While the combined antiretroviral therapy has resulted in a significant decrease in HIV-1 related morbidity and mortality, the HIV-1 pandemic has not been substantially averted. To curtail the 2.4 million new infections each year, a prophylactic HIV-1 vaccine is urgently needed. This review first summarizes four major completed clinical efficacy trials of prophylactic HIV-1 vaccine and their outcomes. Next, it discusses several other approaches that have not yet advanced to clinical efficacy trials, but provided valuable insights into vaccine design. Among them, live-attenuated vaccines (LAVs) provided excellent protection in a non-human primate model. However, safety concerns have precluded the current version of LAVs from clinical application. As the major component of this review, two synthetic biology approaches for improving the safety of HIV-1 LAVs through controlling HIV-1 replication are discussed. Particular focus is on a novel approach that uses unnatural amino acid-mediated suppression of amber nonsense codon to generate conditionally replicating HIV-1 variants. The objective is to attract more attention towards this promising research field and to provoke creative designs and innovative utilization of the two control strategies.
Collapse
Affiliation(s)
- Nanxi Wang
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Zhe Yuan
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Wei Niu
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Qingsheng Li
- Nebraska Center for Virology & School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588, USA
| |
Collapse
|
44
|
Lee S, Sohn KC, Choi DK, Won M, Park KA, Ju SK, Kang K, Bae YK, Hur GM, Ro H. Ecdysone Receptor-based Singular Gene Switches for Regulated Transgene Expression in Cells and Adult Rodent Tissues. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e367. [PMID: 27673563 PMCID: PMC5056996 DOI: 10.1038/mtna.2016.74] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 07/25/2016] [Indexed: 11/09/2022]
Abstract
Controlled gene expression is an indispensable technique in biomedical research. Here, we report a convenient, straightforward, and reliable way to induce expression of a gene of interest with negligible background expression compared to the most widely used tetracycline (Tet)-regulated system. Exploiting a Drosophila ecdysone receptor (EcR)-based gene regulatory system, we generated nonviral and adenoviral singular vectors designated as pEUI(+) and pENTR-EUI, respectively, which contain all the required elements to guarantee regulated transgene expression (GAL4-miniVP16-EcR, termed GvEcR hereafter, and 10 tandem repeats of an upstream activation sequence promoter followed by a multiple cloning site). Through the transient and stable transfection of mammalian cell lines with reporter genes, we validated that tebufenozide, an ecdysone agonist, reversibly induced gene expression, in a dose- and time-dependent manner, with negligible background expression. In addition, we created an adenovirus derived from the pENTR-EUI vector that readily infected not only cultured cells but also rodent tissues and was sensitive to tebufenozide treatment for regulated transgene expression. These results suggest that EcR-based singular gene regulatory switches would be convenient tools for the induction of gene expression in cells and tissues in a tightly controlled fashion.
Collapse
Affiliation(s)
- Seoghyun Lee
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Kyung-Cheol Sohn
- Department of Dermatology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Dae-Kyoung Choi
- Department of Dermatology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Minho Won
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kyeong Ah Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung-Kyu Ju
- Affiliated Research (and Development) Institute, Daejeon, Republic of Korea
| | - Kidong Kang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Young-Ki Bae
- Comparative Biomedical Research Branch, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
45
|
Bizzarri M, Cucina A. SMT and TOFT: Why and How They are Opposite and Incompatible Paradigms. Acta Biotheor 2016; 64:221-39. [PMID: 27283400 DOI: 10.1007/s10441-016-9281-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 05/23/2016] [Indexed: 01/02/2023]
Abstract
The Somatic Mutation Theory (SMT) has been challenged on its fundamentals by the Tissue Organization Field Theory of Carcinogenesis (TOFT). However, a recent publication has questioned whether TOFT could be a valid alternative theory of carcinogenesis to that presented by SMT. Herein we critically review arguments supporting the irreducible opposition between the two theoretical approaches by highlighting differences regarding the philosophical, methodological and experimental approaches on which they respectively rely. We conclude that SMT has not explained carcinogenesis due to severe epistemological and empirical shortcomings, while TOFT is gaining momentum. The main issue is actually to submit SMT to rigorous testing. This concern includes the imperatives to seek evidence for disproving one's hypothesis, and to consider the whole, and not just selective evidence.
Collapse
Affiliation(s)
- Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
- Systems Biology Group Lab, Sapienza University of Rome, Via Antonio Scarpa 14, 00161, Rome, Italy.
| | - Alessandra Cucina
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161, Rome, Italy
- Azienda Policlinico Umberto I, Viale del Policlinico 155, 00161, Rome, Italy
| |
Collapse
|
46
|
rRNA Binding Sites and the Molecular Mechanism of Action of the Tetracyclines. Antimicrob Agents Chemother 2016; 60:4433-41. [PMID: 27246781 DOI: 10.1128/aac.00594-16] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The tetracycline antibiotics are known to be effective in the treatment of both infectious and noninfectious disease conditions. The 16S rRNA binding mechanism currently held for the antibacterial action of the tetracyclines does not explain their activity against viruses, protozoa that lack mitochondria, and noninfectious conditions. Also, the mechanism by which the tetracyclines selectively inhibit microbial protein synthesis against host eukaryotic protein synthesis despite conservation of ribosome structure and functions is still questionable. Many studies have investigated the binding of the tetracyclines to the 16S rRNA using the small ribosomal subunit of different bacterial species, but there seems to be no agreement between various reports on the exact binding site on the 16S rRNA. The wide range of activity of the tetracyclines against a broad spectrum of bacterial pathogens, viruses, protozoa, and helminths, as well as noninfectious conditions, indicates a more generalized effect on RNA. In the light of recent evidence that the tetracyclines bind to various synthetic double-stranded RNAs (dsRNAs) of random base sequences, suggesting that the double-stranded structures may play a more important role in the binding of the tetracyclines to RNA than the specific base pairs, as earlier speculated, it is imperative to consider possible alternative binding modes or sites that could help explain the mechanisms of action of the tetracyclines against various pathogens and disease conditions.
Collapse
|
47
|
Jeong DU, Choi JY, Kim DW. Cartilage-Specific and Cre-Dependent Nkx3.2 Overexpression In Vivo Causes Skeletal Dwarfism by Delaying Cartilage Hypertrophy. J Cell Physiol 2016; 232:78-90. [DOI: 10.1002/jcp.25446] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/01/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Da-Un Jeong
- Department of Biochemistry; College of Life Science and Biotechnology; Yonsei University; Seoul Republic of Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology; School of Medicine, Kyungpook National University; Daegu Republic of Korea
| | - Dae-Won Kim
- Department of Biochemistry; College of Life Science and Biotechnology; Yonsei University; Seoul Republic of Korea
| |
Collapse
|
48
|
Zhang Y, Chen Y, Fu Y, Ying C, Feng Y, Huang Q, Wang C, Pei DS, Wang D. Monitoring tetracycline through a solid-state nanopore sensor. Sci Rep 2016; 6:27959. [PMID: 27306259 PMCID: PMC4910080 DOI: 10.1038/srep27959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/27/2016] [Indexed: 11/29/2022] Open
Abstract
Antibiotics as emerging environmental contaminants, are widely used in both human and veterinary medicines. A solid-state nanopore sensing method is reported in this article to detect Tetracycline, which is based on Tet-off and Tet-on systems. rtTA (reverse tetracycline-controlled trans-activator) and TRE (Tetracycline Responsive Element) could bind each other under the action of Tetracycline to form one complex. When the complex passes through nanopores with 8 ~ 9 nanometers in diameter, we could detect the concentrations of Tet from 2 ng/mL to 2000 ng/mL. According to the Logistic model, we could define three growth zones of Tetracycline for rtTA and TRE. The slow growth zone is 0–39.5 ng/mL. The rapid growth zone is 39.5−529.7 ng/mL. The saturated zone is > 529.7 ng/mL. Compared to the previous methods, the nanopore sensor could detect and quantify these different kinds of molecule at the single-molecule level.
Collapse
Affiliation(s)
- Yuechuan Zhang
- School of Physical Electronic, University of Electronic Science and Technology of China, Chengdu, P.R. China.,Chongqing Key Lab of Multi-scale Manufacturing Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| | - Yanling Chen
- Research Center for Environment and Health, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| | - Yongqi Fu
- School of Physical Electronic, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Cuifeng Ying
- Chongqing Key Lab of Multi-scale Manufacturing Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China.,Key Laboratory of Weak-Light Nonlinear Photonics, Ministry of Education, School of Physics, Nankai University, Tianjin, China
| | - Yanxiao Feng
- Chongqing Key Lab of Multi-scale Manufacturing Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| | - Qimeng Huang
- Chongqing Key Lab of Multi-scale Manufacturing Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| | - Chao Wang
- Research Center for Environment and Health, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| | - De-Sheng Pei
- Research Center for Environment and Health, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| | - Deqiang Wang
- Chongqing Key Lab of Multi-scale Manufacturing Technology, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, P.R. China
| |
Collapse
|
49
|
Möller T, Bard F, Bhattacharya A, Biber K, Campbell B, Dale E, Eder C, Gan L, Garden GA, Hughes ZA, Pearse DD, Staal RGW, Sayed FA, Wes PD, Boddeke HWGM. Critical data-based re-evaluation of minocycline as a putative specific microglia inhibitor. Glia 2016; 64:1788-94. [PMID: 27246804 DOI: 10.1002/glia.23007] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/04/2016] [Indexed: 12/11/2022]
Abstract
Minocycline, a second generation broad-spectrum antibiotic, has been frequently postulated to be a "microglia inhibitor." A considerable number of publications have used minocycline as a tool and concluded, after achieving a pharmacological effect, that the effect must be due to "inhibition" of microglia. It is, however, unclear how this "inhibition" is achieved at the molecular and cellular levels. Here, we weigh the evidence whether minocycline is indeed a bona fide microglia inhibitor and discuss how data generated with minocycline should be interpreted. GLIA 2016;64:1788-1794.
Collapse
Affiliation(s)
- Thomas Möller
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey.,Department of Neurology, University of Washington, Seattle, Washington
| | | | - Anindya Bhattacharya
- Janssen Research & Development, LLC., Neuroscience Drug Discovery, San Diego, California
| | - Knut Biber
- Department of Psychiatry and Psychotherapy, University Hospital Freiburg, Freiburg, Germany.,Department of Neuroscience, University Medical Center Groningen, Groningen, The Netherlands
| | - Brian Campbell
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Elena Dale
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Claudia Eder
- Institute for Infection and Immunity, St. George's - University of London, London, United Kingdom
| | - Li Gan
- Gladstone Institute for Neurodegeneration, San Francisco, California
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, Washington
| | - Zoë A Hughes
- Neuroscience & Pain Research Unit, Pfizer Global Research, Cambridge, Massachusetts
| | - Damien D Pearse
- Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Roland G W Staal
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Faten A Sayed
- Gladstone Institute for Neurodegeneration, San Francisco, California.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, California
| | - Paul D Wes
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | | |
Collapse
|
50
|
Wang JL, Huang SY, Behnke MS, Chen K, Shen B, Zhu XQ. The Past, Present, and Future of Genetic Manipulation in Toxoplasma gondii. Trends Parasitol 2016; 32:542-553. [PMID: 27184069 DOI: 10.1016/j.pt.2016.04.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/16/2022]
Abstract
Toxoplasma gondii is a classic model for studying obligate intracellular microorganisms as various genetic manipulation tools have been developed in T. gondii over the past 20 years. Here we summarize the major strategies for T. gondii genetic manipulation including genetic crosses, insertional mutagenesis, chemical mutagenesis, homologous gene replacement, conditional knockdown techniques, and the recently developed clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system. We evaluate the advantages and limitations of each of these tools in a historical perspective. We also discuss additional applications of modified CRISPR-Cas9 systems for use in T. gondii, such as regulation of gene expression, labeling of specific genomic loci, and epigenetic modifications. These approaches have the potential to revolutionize the analysis of T. gondii biology and help us to better develop new drugs and vaccines.
Collapse
Affiliation(s)
- Jin-Lei Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Si-Yang Huang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province 225009, PR China
| | - Michael S Behnke
- Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Kai Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China.
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province 225009, PR China.
| |
Collapse
|