101
|
Sawada T, Itoh M, Nakamura Y. Contributions of Three Starch Branching Enzyme Isozymes to the Fine Structure of Amylopectin in Rice Endosperm. FRONTIERS IN PLANT SCIENCE 2018; 9:1536. [PMID: 30405671 PMCID: PMC6206275 DOI: 10.3389/fpls.2018.01536] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/28/2018] [Indexed: 05/07/2023]
Abstract
Three starch branching enzyme (BE) isozymes, BEI, BEIIa, and BEIIb, are involved in starch biosynthesis in rice endosperm. Past in vivo and in vitro studies have suggested that each BE isozyme plays a distinct role in forming the fine structure of amylopectin. To elucidate more details of their roles, we prepared DNA constructs in which all the possible combinations of the expressions of these three isozymes were suppressed in developing rice endosperm. Analysis of the chain-length distributions of amylopectin produced under these various conditions confirmed the contributions of the individual BE isozymes to the fine structure of amylopectin in rice endosperm. Among these isozymes, the impact of loss of BEIIb activity on amylopectin fine structure was most remarkable and indicated that it plays a specific role in the synthesis of short chains with a 6-13 degree of polymerization (DP). The contribution of BEI to the amylopectin synthesis was unclear when only BEI activity was reduced. It was clear, however, when both BEI and BEIIb activities were substantially inhibited. The DP11-22 intermediate chains were markedly reduced in the ΔBEI/BEIIb line compared with the ΔBEIIb line, indicating that BEI plays a distinct role in the synthesis of these intermediate chains. Although no substantial change in amylopectin chain profile was detected in the ΔBEIIa line, the role of BEIIa could be deciphered by analyzing amylopectin fine structure from the ΔBEI/BEIIa/BEIIb line in comparison to that from ΔBEI/BEIIb line. This strongly suggests that BEIIa compensates for the role of BEI, rather than that of BEIIb, by forming intermediate chains of DP11-22. In addition, the new possibility that BEIIa is involved in the formation of starch granules in rice endosperm was suggested because the onset temperature for gelatinization of starch granules in the ΔBEIIa/BEIIb line was significantly higher than that in the ΔBEIIb line. In summary, the present study highlights the distinct roles of BEI, BEIIa, and BEIIb in the synthesis of amylopectin in developing rice endosperm.
Collapse
Affiliation(s)
- Takayuki Sawada
- Faculty of Bioresource Sciences, Akita Prefectural University, Akita, Japan
| | - Mizuho Itoh
- Faculty of Bioresource Sciences, Akita Prefectural University, Akita, Japan
| | - Yasunori Nakamura
- Faculty of Bioresource Sciences, Akita Prefectural University, Akita, Japan
- Akita Natural Science Laboratory, Akita, Japan
| |
Collapse
|
102
|
Fischer MD, Mgboji E, Liu Z. Pyrite cloning: a single tube and programmed reaction cloning with restriction enzymes. PLANT METHODS 2018; 14:91. [PMID: 30349582 PMCID: PMC6192209 DOI: 10.1186/s13007-018-0359-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/10/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Insertion of engineered DNA fragments into bacterial vectors is the foundation of recombinant DNA technology, yet existing methods are still laborious, require many steps, depend on specific vector configuration, or require expensive reagents. RESULTS We have developed a method, called "Pyrite" cloning that combines the traditional restriction enzyme digestion and ligation reaction in a single tube and uses a programmed thermocycler reaction, allowing rapid and flexible cloning in a single tube. After the Pyrite reaction and transformation, approximately 50% colonies contain the expected insert, which can be easily and quickly determined by colony PCR or blue-white colony screening. We also demonstrated that Pyrite cloning can be applied for different cloning purposes. CONCLUSIONS The Pyrite cloning method reported here is a single tube and programmed reaction cloning with restriction enzymes. Compared to other cloning methods, Pyrite cloning is flexible, inexpensive, simple, and highly efficient.
Collapse
Affiliation(s)
- Matthew D. Fischer
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Emmanuel Mgboji
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Zhongchi Liu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
103
|
Guidelli GV, Mattiello L, Gallinari RH, Lucca PCD, Menossi M. pGVG: a new Gateway-compatible vector for transformation of sugarcane and other monocot crops. Genet Mol Biol 2018; 41:450-454. [PMID: 30088611 PMCID: PMC6082244 DOI: 10.1590/1678-4685-gmb-2017-0262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/27/2017] [Indexed: 11/21/2022] Open
Abstract
The successful development of genetically engineered monocots using Agrobacterium-mediated transformation has created an increasing demand for compatible vectors. We have developed a new expression vector, pGVG, for efficient transformation and expression of different constructs for gene overexpression and silencing in sugarcane. The pCAMBIA2300 binary vector was modified by adding Gateway recombination sites for fast gene transfer between vectors and the maize polyubiquitin promoter Ubi-1 (ZmUbi1), which is known to drive high gene expression levels in monocots. Transformation efficiency using the pGVG vector reached up to 14 transgenic events per gram of transformed callus. Transgenic plants expressing the β-glucuronidase (GUS) reporter gene from pGVG showed high levels of GUS activity. qRT-PCR evaluations demonstrated success for both overexpression and hairpin-based silencing cassettes. Therefore, pGVG is suitable for plant transformation and subsequent applications for high-throughput production of stable transgenic sugarcane. The use of an expression cassette based on the ZmUbi1 promoter opens the possibility of using pGVG in other monocot species.
Collapse
Affiliation(s)
- Giovanna V Guidelli
- Laboratório de Genoma Funcional, Universidade Estadual de Campinas, Departamento de Genética, Evolução e Bioagentes, Instituto de Biologia, Campinas, SP, Brazil
| | - Lucia Mattiello
- Laboratório de Genoma Funcional, Universidade Estadual de Campinas, Departamento de Genética, Evolução e Bioagentes, Instituto de Biologia, Campinas, SP, Brazil
| | - Rafael H Gallinari
- Laboratório de Genoma Funcional, Universidade Estadual de Campinas, Departamento de Genética, Evolução e Bioagentes, Instituto de Biologia, Campinas, SP, Brazil
| | - Paulo Cezar de Lucca
- PangeiaBiotech, Universidade Estadual de Campinas, Departamento de Genética, Evolução, Microbiologia e Imunologia, Instituto de Biologia, Campinas, SP, Brazil
| | - Marcelo Menossi
- Laboratório de Genoma Funcional, Universidade Estadual de Campinas, Departamento de Genética, Evolução e Bioagentes, Instituto de Biologia, Campinas, SP, Brazil
| |
Collapse
|
104
|
Pouvreau B, Vanhercke T, Singh S. From plant metabolic engineering to plant synthetic biology: The evolution of the design/build/test/learn cycle. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2018; 273:3-12. [PMID: 29907306 DOI: 10.1016/j.plantsci.2018.03.035] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/19/2018] [Accepted: 03/28/2018] [Indexed: 05/21/2023]
Abstract
Genetic improvement of crops started since the dawn of agriculture and has continuously evolved in parallel with emerging technological innovations. The use of genome engineering in crop improvement has already revolutionised modern agriculture in less than thirty years. Plant metabolic engineering is still at a development stage and faces several challenges, in particular with the time necessary to develop plant based solutions to bio-industrial demands. However the recent success of several metabolic engineering approaches applied to major crops are encouraging and the emerging field of plant synthetic biology offers new opportunities. Some pioneering studies have demonstrated that synthetic genetic circuits or orthogonal metabolic pathways can be introduced into plants to achieve a desired function. The combination of metabolic engineering and synthetic biology is expected to significantly accelerate crop improvement. A defining aspect of both fields is the design/build/test/learn cycle, or the use of iterative rounds of testing modifications to refine hypotheses and develop best solutions. Several technological and technical improvements are now available to make a better use of each design, build, test, and learn components of the cycle. All these advances should facilitate the rapid development of a wide variety of bio-products for a world in need of sustainable solutions.
Collapse
Affiliation(s)
- Benjamin Pouvreau
- CSIRO Agriculture and Food, PO Box 1600, Canberra, ACT 2601, Australia.
| | - Thomas Vanhercke
- CSIRO Agriculture and Food, PO Box 1600, Canberra, ACT 2601, Australia
| | - Surinder Singh
- CSIRO Agriculture and Food, PO Box 1600, Canberra, ACT 2601, Australia
| |
Collapse
|
105
|
Arévalo-Pinzón G, González-González M, Suárez CF, Curtidor H, Carabias-Sánchez J, Muro A, LaBaer J, Patarroyo MA, Fuentes M. Self-assembling functional programmable protein array for studying protein-protein interactions in malaria parasites. Malar J 2018; 17:270. [PMID: 30016987 PMCID: PMC6050706 DOI: 10.1186/s12936-018-2414-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/10/2018] [Indexed: 12/30/2022] Open
Abstract
Background Plasmodium vivax is the most widespread malarial species, causing significant morbidity worldwide. Knowledge is limited regarding the molecular mechanism of invasion due to the lack of a continuous in vitro culture system for these species. Since protein–protein and host–cell interactions play an essential role in the microorganism’s invasion and replication, elucidating protein function during invasion is critical when developing more effective control methods. Nucleic acid programmable protein array (NAPPA) has thus become a suitable technology for studying protein–protein and host–protein interactions since producing proteins through the in vitro transcription/translation (IVTT) method overcomes most of the drawbacks encountered to date, such as heterologous protein production, stability and purification. Results Twenty P. vivax proteins on merozoite surface or in secretory organelles were selected and successfully cloned using gateway technology. Most constructs were displayed in the array expressed in situ, using the IVTT method. The Pv12 protein was used as bait for evaluating array functionality and co-expressed with P. vivax cDNA display in the array. It was found that Pv12 interacted with Pv41 (as previously described), as well as PvMSP142kDa, PvRBP1a, PvMSP8 and PvRAP1. Conclusions NAPPA is a high-performance technique enabling co-expression of bait and query in situ, thereby enabling interactions to be analysed rapidly and reproducibly. It offers a fresh alternative for studying protein–protein and ligand–receptor interactions regarding a parasite which is difficult to cultivate (i.e. P. vivax). Electronic supplementary material The online version of this article (10.1186/s12936-018-2414-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Arévalo-Pinzón
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | - María González-González
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.,Department of Medicine and General Cytometry Service-Nucleus, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain
| | - Carlos Fernando Suárez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 # 55-37, Bogotá, Colombia
| | - Hernando Curtidor
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | | | - Antonio Muro
- Unidad de Investigación Enfermedades Infecciosas y Tropicales (e-INTRO), Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (IBSAL-CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Universitario Miguel de Unamuno s/n, 37007, Salamanca, Spain
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | - Manuel Fuentes
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain. .,Department of Medicine and General Cytometry Service-Nucleus, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
106
|
Samavarchi-Tehrani P, Abdouni H, Samson R, Gingras AC. A Versatile Lentiviral Delivery Toolkit for Proximity-dependent Biotinylation in Diverse Cell Types. Mol Cell Proteomics 2018; 17:2256-2269. [PMID: 29991506 DOI: 10.1074/mcp.tir118.000902] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Indexed: 11/06/2022] Open
Abstract
Proximity-dependent biotinylation strategies have emerged as powerful tools to characterize the subcellular context of proteins in living cells. The popular BioID approach employs an abortive E. coli biotin ligase mutant (R118G; denoted as BirA*), which when fused to a bait protein enables the covalent biotinylation of endogenous proximal polypeptides. This approach has been mainly applied to the study of protein proximity in immortalized mammalian cell lines. To expand the application space of BioID, here we describe a set of lentiviral vectors that enable the inducible expression of BirA*-tagged bait fusion proteins for performing proximity-dependent biotinylation in diverse experimental systems. We benchmark this highly adaptable toolkit across immortalized and primary cell systems, demonstrating the ease, versatility and robustness of the system. We also provide guidelines to perform BioID using these reagents.
Collapse
Affiliation(s)
| | - Hala Abdouni
- From the ‡Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Reuben Samson
- From the ‡Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada.,§Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Anne-Claude Gingras
- From the ‡Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada; .,§Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
107
|
Survey of Human Chromosome 21 Gene Expression Effects on Early Development in Danio rerio. G3-GENES GENOMES GENETICS 2018; 8:2215-2223. [PMID: 29760202 PMCID: PMC6027891 DOI: 10.1534/g3.118.200144] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Trisomy for human chromosome 21 (Hsa21) results in Down syndrome (DS), one of the most genetically complex conditions compatible with human survival. Assessment of the physiological consequences of dosage-driven overexpression of individual Hsa21 genes during early embryogenesis and the resulting contributions to DS pathology in mammals are not tractable in a systematic way. A recent study looked at loss-of-function of a subset of Caenorhabditis elegans orthologs of Hsa21 genes and identified ten candidates with behavioral phenotypes, but the equivalent over-expression experiment has not been done. We turned to zebrafish as a developmental model and, using a number of surrogate phenotypes, we screened Hsa21 genes for effects on early embyrogenesis. We prepared a library of 164 cDNAs of conserved protein coding genes, injected mRNA into early embryos and evaluated up to 5 days post-fertilization (dpf). Twenty-four genes produced a gross morphological phenotype, 11 of which could be reproduced reliably. Seven of these gave a phenotype consistent with down regulation of the sonic hedgehog (Shh) pathway; two showed defects indicative of defective neural crest migration; one resulted consistently in pericardial edema; and one was embryonic lethal. Combinatorial injections of multiple Hsa21 genes revealed both additive and compensatory effects, supporting the notion that complex genetic relationships underlie end phenotypes of trisomy that produce DS. Together, our data suggest that this system is useful in the genetic dissection of dosage-sensitive gene effects on early development and can inform the contribution of both individual loci and their combinatorial effects to phenotypes relevant to the etiopathology of DS.
Collapse
|
108
|
Yu D, Liao L, Zhang Y, Xu K, Zhang J, Liu K, Li X, Tan G, Zheng J, He Y, Xu C, Zhao J, Fu B, Xie J, Mao J, Li C. Development of a Gateway-compatible pCAMBIA binary vector for RNAi-mediated gene knockdown in plants. Plasmid 2018; 98:52-55. [PMID: 30201136 DOI: 10.1016/j.plasmid.2018.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/25/2018] [Accepted: 09/05/2018] [Indexed: 11/24/2022]
Abstract
RNA interference (RNAi), based on hairpin RNA (hpRNA) expression, plays an important role in functional analysis of plant genes. Traditional methods for making RNAi constructs usually involve multiple time-consuming cloning steps. We have developed a Gateway-compatible binary vector for RNAi-mediated gene knockdown in plants from pCAMBIA2301 and pHANNIBAL vectors. The new plant RNAi binary vector, named pCAMBIA2301-GW-RNAi, has two inverted repeated Gateway cassettes driven by the cauliflower mosaic virus 35S (CaMV 35S) promoter. This enables site-specific recombination at two sites by one Gateway LR reaction without restriction enzymes and ligases. The pCAMBIA2301-GW-RNAi vector's effectiveness was evaluated by Agrobacterium-mediated transient co-expression assays of overexpression and silencing constructs of HvCEBiP in Nicotiana benthamiana followed by western blot analysis. Obtained results show that the developed RNAi vector successfully knocked down 35S-driven expression of HvCEBiP, as expression levels of the encoded HvCEBiP protein were significantly reduced.
Collapse
Affiliation(s)
- Deshui Yu
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China
| | - Libing Liao
- Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Yi Zhang
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Agronomy, Henan Agricultural University, Zhengzhou 450002, China
| | - Kedong Xu
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China
| | - Ju Zhang
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China
| | - Kun Liu
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China
| | - Xiaoli Li
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China
| | - Guangxuan Tan
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China
| | - Jurui Zheng
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Yong He
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Changling Xu
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Jinjin Zhao
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Beibei Fu
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Jiaxing Xie
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Jie Mao
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, China
| | - Chengwei Li
- Key Laboratory of Plant Genetics and Molecular Breeding, Zhoukou Normal University, Zhoukou 466001, China; Henan Key Laboratory of Crop Molecular Breeding & Bioreactor, Zhoukou 466001, China; Henan Engineering Research Center of Crop Genome Editing, Henan Institute of Science and Technology, Xinxiang 453003, China.
| |
Collapse
|
109
|
Gantner J, Ordon J, Ilse T, Kretschmer C, Gruetzner R, Löfke C, Dagdas Y, Bürstenbinder K, Marillonnet S, Stuttmann J. Peripheral infrastructure vectors and an extended set of plant parts for the Modular Cloning system. PLoS One 2018; 13:e0197185. [PMID: 29847550 PMCID: PMC5976141 DOI: 10.1371/journal.pone.0197185] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/27/2018] [Indexed: 11/21/2022] Open
Abstract
Standardized DNA assembly strategies facilitate the generation of multigene constructs from collections of building blocks in plant synthetic biology. A common syntax for hierarchical DNA assembly following the Golden Gate principle employing Type IIs restriction endonucleases was recently developed, and underlies the Modular Cloning and GoldenBraid systems. In these systems, transcriptional units and/or multigene constructs are assembled from libraries of standardized building blocks, also referred to as phytobricks, in several hierarchical levels and by iterative Golden Gate reactions. Here, a toolkit containing further modules for the novel DNA assembly standards was developed. Intended for use with Modular Cloning, most modules are also compatible with GoldenBraid. Firstly, a collection of approximately 80 additional phytobricks is provided, comprising e.g. modules for inducible expression systems, promoters or epitope tags. Furthermore, DNA modules were developed for connecting Modular Cloning and Gateway cloning, either for toggling between systems or for standardized Gateway destination vector assembly. Finally, first instances of a "peripheral infrastructure" around Modular Cloning are presented: While available toolkits are designed for the assembly of plant transformation constructs, vectors were created to also use coding sequence-containing phytobricks directly in yeast two hybrid interaction or bacterial infection assays. The presented material will further enhance versatility of hierarchical DNA assembly strategies.
Collapse
Affiliation(s)
- Johannes Gantner
- Institute for Biology, Department of Plant Genetics, Martin Luther University Halle (Saale), Halle, Germany
| | - Jana Ordon
- Institute for Biology, Department of Plant Genetics, Martin Luther University Halle (Saale), Halle, Germany
| | - Theresa Ilse
- Institute for Biology, Department of Plant Genetics, Martin Luther University Halle (Saale), Halle, Germany
| | - Carola Kretschmer
- Institute for Biology, Department of Plant Genetics, Martin Luther University Halle (Saale), Halle, Germany
| | - Ramona Gruetzner
- Department of Cell and Metabolic Biology, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Christian Löfke
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna BioCenter (VBC), Vienna, Austria
| | - Yasin Dagdas
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna BioCenter (VBC), Vienna, Austria
| | - Katharina Bürstenbinder
- Department of Molecular Signal Processing, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Sylvestre Marillonnet
- Department of Cell and Metabolic Biology, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Johannes Stuttmann
- Institute for Biology, Department of Plant Genetics, Martin Luther University Halle (Saale), Halle, Germany
| |
Collapse
|
110
|
A novel, easy and rapid method for constructing yeast two-hybrid vectors using In-Fusion technology. Biotechniques 2018; 64:219-224. [PMID: 29673256 DOI: 10.2144/btn-2018-0007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Yeast two-hybrid systems are powerful tools for analyzing interactions between proteins. Vector construction is an essential step in yeast two-hybrid experiments, which require bait and prey plasmids. In this study, we modified the multiple cloning site sequence of the yeast plasmid pGADT7 by site-directed mutagenesis PCR to generate the pGADT7-In vector, which resulted in an easy and rapid method for constructing yeast two-hybrid vectors using the In-Fusion cloning technique. This method has three key advantages: only one pair of primers and one round of PCR are needed to generate bait and prey plasmids for each gene, it is restriction endonuclease- and ligase-independent, and it is fast and easily performed.
Collapse
|
111
|
Perry EB, Makohon-Moore A, Zheng C, Kaufman CK, Cai J, Iacobuzio-Donahue CA, White RM. Tumor diversity and evolution revealed through RADseq. Oncotarget 2018; 8:41792-41805. [PMID: 28611298 PMCID: PMC5522028 DOI: 10.18632/oncotarget.18355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 05/12/2017] [Indexed: 12/30/2022] Open
Abstract
Cancer is an evolutionary disease, and there is increasing interest in applying tools from evolutionary biology to understand cancer progression. Restriction-site associated DNA sequencing (RADseq) was developed for the field of evolutionary genetics to study adaptation and identify evolutionary relationships among populations. Here we apply RADseq to study tumor evolution, which allows for unbiased sampling of any desired frequency of the genome, overcoming the selection bias and cost limitations inherent to exome or whole-genome sequencing. We apply RADseq to both human pancreatic cancer and zebrafish melanoma samples. Using either a low-frequency (SbfI, 0.4% of the genome) or high-frequency (NsiI, 6-9% of the genome) cutter, we successfully identify single nucleotide substitutions and copy number alterations in tumors, which can be augmented by performing RADseq on sublineages within the tumor. We are able to infer phylogenetic relationships between primary tumors and metastases. These same methods can be used to identify somatic mosaicism in seemingly normal, non-cancerous tissues. Evolutionary studies of cancer that focus on rates of tumor evolution and evolutionary relationships among tumor lineages will benefit from the flexibility and efficiency of restriction-site associated DNA sequencing.
Collapse
Affiliation(s)
- Elizabeth B Perry
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Biostatistics, Yale University, New Haven, Connecticut, USA
| | - Alvin Makohon-Moore
- The David M. Rubenstein Center for Pancreatic Cancer Research, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Caihong Zheng
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Jun Cai
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Christine A Iacobuzio-Donahue
- The David M. Rubenstein Center for Pancreatic Cancer Research, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Richard M White
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
112
|
Lai YT, Deem KD, Borràs-Castells F, Sambrani N, Rudolf H, Suryamohan K, El-Sherif E, Halfon MS, McKay DJ, Tomoyasu Y. Enhancer identification and activity evaluation in the red flour beetle, Tribolium castaneum. Development 2018; 145:dev160663. [PMID: 29540499 PMCID: PMC11736658 DOI: 10.1242/dev.160663] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/09/2018] [Indexed: 12/13/2022]
Abstract
Evolution of cis-regulatory elements (such as enhancers) plays an important role in the production of diverse morphology. However, a mechanistic understanding is often limited by the absence of methods for studying enhancers in species other than established model systems. Here, we sought to establish methods to identify and test enhancer activity in the red flour beetle, Tribolium castaneum To identify possible enhancer regions, we first obtained genome-wide chromatin profiles from various tissues and stages of Tribolium using FAIRE (formaldehyde-assisted isolation of regulatory elements)-sequencing. Comparison of these profiles revealed a distinct set of open chromatin regions in each tissue and at each stage. In addition, comparison of the FAIRE data with sets of computationally predicted (i.e. supervised cis-regulatory module-predicted) enhancers revealed a very high overlap between the two datasets. Second, using nubbin in the wing and hunchback in the embryo as case studies, we established the first universal reporter assay system that works in various contexts in Tribolium, and in a cross-species context. Together, these advances will facilitate investigation of cis-evolution and morphological diversity in Tribolium and other insects.
Collapse
Affiliation(s)
- Yi-Ting Lai
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Kevin D Deem
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | | | - Nagraj Sambrani
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Heike Rudolf
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Kushal Suryamohan
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Ezzat El-Sherif
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Marc S Halfon
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Daniel J McKay
- Department of Biology, Department of Genetics, Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
113
|
Movahedi K, Wiegmann R, De Vlaminck K, Van Ginderachter JA, Nikolaev VO. RoMo: An efficient strategy for functional mosaic analysis via stochastic Cre recombination and gene targeting in theROSA26locus. Biotechnol Bioeng 2018; 115:1778-1792. [DOI: 10.1002/bit.26594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/20/2018] [Accepted: 03/15/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Kiavash Movahedi
- Myeloid Cell Immunology Lab; VIB Center for Inflammation Research; Brussels Belgium
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels Belgium
- Max Planck Institute of Biophysics; Max-von-Laue-Strasse 3; Frankfurt Germany
| | - Robert Wiegmann
- Institute of Experimental Cardiovascular Research, University Medical Hamburg-Eppendorf, DZHK (German Center for Cardiovascular Research); Partner Site Hamburg/Kiel/Lübeck; Hamburg Germany
| | - Karen De Vlaminck
- Myeloid Cell Immunology Lab; VIB Center for Inflammation Research; Brussels Belgium
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels Belgium
| | - Jo A. Van Ginderachter
- Myeloid Cell Immunology Lab; VIB Center for Inflammation Research; Brussels Belgium
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels Belgium
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Hamburg-Eppendorf, DZHK (German Center for Cardiovascular Research); Partner Site Hamburg/Kiel/Lübeck; Hamburg Germany
| |
Collapse
|
114
|
Hubner EK, Lechler C, Kohnke-Ertel B, Zmoos AF, Sage J, Schmid RM, Ehmer U. An in vivo transfection system for inducible gene expression and gene silencing in murine hepatocytes. J Gene Med 2018; 19. [PMID: 28009940 DOI: 10.1002/jgm.2940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hydrodynamic tail vein injection (HTVI) of transposon-based integration vectors is an established system for stably transfecting mouse hepatocytes in vivo that has been successfully employed to study key questions in liver biology and cancer. Refining the vectors for transposon-mediated hepatocyte transfection will further expand the range of applications of this technique in liver research. In the present study, we report an advanced transposon-based system for manipulating gene expression in hepatocytes in vivo. METHODS Transposon-based vector constructs were generated to enable the constitutive expression of inducible Cre recombinase (CreER) together with tetracycline-inducible transgene or miR-small hairpin RNA (shRNA) expression (Tet-ON system). Transposon and transposase expression vectors were co-injected into R26R-mTmG reporter mice by HTVI. Cre-mediated gene recombination was induced by tamoxifen, followed by the administration of doxycycline to drive tetracycline-inducible gene or shRNA expression. Expression was visualized by immunofluorescence staining in livers of injected mice. RESULTS After HTVI, Cre recombination by tamoxifen led to the expression of membrane-bound green fluorescent protein in transfected hepatocytes. Activation of inducible gene or shRNA expression was detected by immunostaining in up to one-third of transfected hepatocytes, with an efficiency dependent on the promoter driving the Tet-ON system. CONCLUSIONS Our vector system combines Cre-lox mediated gene mutation with inducible gene expression or gene knockdown, respectively. It provides the opportunity for rapid and specific modification of hepatocyte gene expression and can be a useful tool for genetic screening approaches and analysis of target genes specifically in genetically engineered mouse models.
Collapse
Affiliation(s)
- Eric K Hubner
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Lechler
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Birgit Kohnke-Ertel
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Anne-Flore Zmoos
- Department of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Julien Sage
- Department of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Roland M Schmid
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ursula Ehmer
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
115
|
Jazayeri SH, Amiri-Yekta A, Bahrami S, Gourabi H, Sanati MH, Khorramizadeh MR. Vector and Cell Line Engineering Technologies Toward Recombinant Protein Expression in Mammalian Cell Lines. Appl Biochem Biotechnol 2018; 185:986-1003. [PMID: 29396733 DOI: 10.1007/s12010-017-2689-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/29/2017] [Indexed: 11/26/2022]
Abstract
The rapid growth of global biopharmaceutical market in the recent years has been a good indication of its significance in biotechnology industry. During a long period of time in recombinant protein production from 1980s, optimizations in both upstream and downstream processes were launched. In this regard, one of the most promising strategies is expression vector engineering technology based on incorporation of DNA opening elements found in the chromatin border regions of vectors as well as targeting gene integration. Along with these approaches, cell line engineering has revealed convenient outcomes in isolating high-producing clones. According to the fact that more than 50% of the approved therapeutic proteins is being manufactured in mammalian cell lines, in this review, we focus on several approaches and developments in vector and cell line engineering technologies in mammalian cell culture.
Collapse
Affiliation(s)
- Seyedeh Hoda Jazayeri
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, P.O. Box: 14155-6343, Tehran, Iran
| | - Amir Amiri-Yekta
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, P.O. Box: 14155-6343, Tehran, Iran
| | - Salahadin Bahrami
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, P.O. Box: 14155-6343, Tehran, Iran
| | - Hamid Gourabi
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, P.O. Box: 14155-6343, Tehran, Iran
| | - Mohammad Hossein Sanati
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, P.O. Box: 14155-6343, Tehran, Iran.
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| | - Mohammad Reza Khorramizadeh
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, P.O. Box: 1411413137, Tehran, Iran.
| |
Collapse
|
116
|
Hubner EK, Lechler C, Rösner TN, Kohnke-Ertel B, Schmid RM, Ehmer U. Constitutive and Inducible Systems for Genetic In Vivo Modification of Mouse Hepatocytes Using Hydrodynamic Tail Vein Injection. J Vis Exp 2018. [PMID: 29443066 DOI: 10.3791/56613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In research models of liver cancer, regeneration, inflammation, and fibrosis, flexible systems for in vivo gene expression and silencing are highly useful. Hydrodynamic tail vein injection of transposon-based constructs is an efficient method for genetic manipulation of hepatocytes in adult mice. In addition to constitutive transgene expression, this system can be used for more advanced applications, such as shRNA-mediated gene knock-down, implication of the CRISPR/Cas9 system to induce gene mutations, or inducible systems. Here, the combination of constitutive CreER expression together with inducible expression of a transgene or miR-shRNA of choice is presented as an example of this technique. We cover the multi-step procedure starting from the preparation of sleeping beauty-transposon constructs, to the injection and treatment of mice, and the preparation of liver tissue for analysis by immunostaining. The system presented is a reliable and efficient approach to achieve complex genetic manipulations in hepatocytes. It is specifically useful in combination with Cre/loxP-based mouse strains and can be applied to a variety of models in the research of liver disease.
Collapse
Affiliation(s)
- Eric K Hubner
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München; Department of Pneumology, Center for Medicine, Medical Center University of Freiburg
| | - Christian Lechler
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München
| | - Thomas N Rösner
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München
| | - Birgit Kohnke-Ertel
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München
| | - Roland M Schmid
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München
| | - Ursula Ehmer
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München;
| |
Collapse
|
117
|
Retraction: Site‐specific recombination of nitrogen‐fixation genes in cyanobacteria by XisF–XisH–XisI complex: Structures and models, William C. Hwang, James W. Golden, Jaime Pascual, Dong Xu, Anton Cheltsov, Adam Godzik. Proteins 2018; 86:268. [PMID: 30338965 PMCID: PMC5094899 DOI: 10.1002/prot.24679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The above article from the Proteins: Structure, Function, and Bioinformatics, published online on 1 September 2014 in Wiley Online Library as Accepted Article (http://onlinelibrary.wiley.com/doi/10.1002/prot.24679/full), has been retracted by agreement between William C. Hwang, James W. Golden, Jaime Pascual, Dong Xu, Anton Cheltsov, Adam Godzik, the Editor‐in‐Chief, Bertrand E. Garcia‐Moreno, and Wiley Periodicals, Inc. The retraction has been agreed because submission was made without agreement from co‐author Adam Godzik.
Collapse
|
118
|
Merentie M, Rissanen R, Lottonen-Raikaslehto L, Huusko J, Gurzeler E, Turunen MP, Holappa L, Mäkinen P, Ylä-Herttuala S. Doxycycline modulates VEGF-A expression: Failure of doxycycline-inducible lentivirus shRNA vector to knockdown VEGF-A expression in transgenic mice. PLoS One 2018; 13:e0190981. [PMID: 29351307 PMCID: PMC5774698 DOI: 10.1371/journal.pone.0190981] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/22/2017] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A) is the master regulator of angiogenesis, vascular permeability and growth. However, its role in mature blood vessels is still not well understood. To better understand the role of VEGF-A in the adult vasculature, we generated a VEGF-A knockdown mouse model carrying a doxycycline (dox)-regulatable short hairpin RNA (shRNA) transgene, which silences VEGF-A. The aim was to find the critical level of VEGF-A reduction for vascular well-being in vivo. In vitro, the dox-inducible lentiviral shRNA vector decreased VEGF-A expression efficiently and dose-dependently in mouse endothelial cells and cardiomyocytes. In the generated transgenic mice plasma VEGF-A levels decreased shortly after the dox treatment but returned back to normal after two weeks. VEGF-A expression decreased shortly after the dox treatment only in some tissues. Surprisingly, increasing the dox exposure time and dose led to elevated VEGF-A expression in some tissues of both wildtype and knockdown mice, suggesting that dox itself has an effect on VEGF-A expression. When the effect of dox on VEGF-A levels was further tested in naïve/non-transduced cells, the dox administration led to a decreased VEGF-A expression in endothelial cells but to an increased expression in cardiomyocytes. In conclusion, the VEGF-A knockdown was achieved in a dox-regulatable fashion with a VEGF-A shRNA vector in vitro, but not in the knockdown mouse model in vivo. Dox itself was found to regulate VEGF-A expression explaining the unexpected results in mice. The effect of dox on VEGF-A levels might at least partly explain its previously reported beneficial effects on myocardial and brain ischemia. Also, this effect on VEGF-A should be taken into account in all studies using dox-regulated vectors.
Collapse
Affiliation(s)
- Mari Merentie
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riina Rissanen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Line Lottonen-Raikaslehto
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jenni Huusko
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Erika Gurzeler
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mikko P. Turunen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lari Holappa
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Petri Mäkinen
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
- Heart Center, Kuopio University Hospital, Kuopio, Finland
- * E-mail:
| |
Collapse
|
119
|
Abstract
Hypoxia has long been recognized as a driving force of tumor progression and therapeutic resistance, and the transcription factor HIF-1α is believed to play a crucial role in these processes. Here we describe an efficient RCAS/Nes-TVA system that allows for in vivo manipulation of HIF-1α expression in the mouse neural progenitor cells. Simple production of the recombinant avian virus RCAS enables quick delivery of gene of interest through injection into the neural progenitors of transgenic mice expressing the viral cognate receptor TVA under the nestin promoter. By crossing with various commercially available genetically engineered mouse strains, a repertoire of mouse models can be created to study gene-specific effects on glioma genesis. This chapter provides details of plasmid construction, viral production, and intracranial delivery of transgenes, a methodology that can be easily adapted to a specific purpose.
Collapse
|
120
|
Zhang Y, Li S, Xue S, Yang S, Huang J, Wang L. Phylogenetic and CRISPR/Cas9 Studies in Deciphering the Evolutionary Trajectory and Phenotypic Impacts of Rice ERECTA Genes. FRONTIERS IN PLANT SCIENCE 2018; 9:473. [PMID: 29692796 PMCID: PMC5902711 DOI: 10.3389/fpls.2018.00473] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 03/26/2018] [Indexed: 05/03/2023]
Abstract
The ERECTA family genes (ERfs) have been found to play diverse functions in Arabidopsis, including controlling cell proliferation and cell growth, regulating stomata patterning, and responding to various stresses. This wide range of functions has rendered them as a potential candidate for crop improvement. However, information on their functional roles, particularly their morphological impact, in crop genomes, such as rice, is limited. Here, through evolutionary prediction, we first depict the evolutionary trajectory of the ER family, and show that the ER family is actually highly conserved across different species, suggesting that most of their functions may also be observed in other plant species. We then take advantage of the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats-associated nuclease 9) system to assess their morphological impact on one of the most important crops, rice. Loss-of-function mutants of OsER1 and OsER2 display shortened plant stature and reduced panicle size, suggesting they possibly also functioned in regulating cell proliferation and cell growth in rice. In addition to functions similar to that in Arabidopsis, we also find clues that rice ERfs may play unique functional roles. The OsER2 displayed more severe phenotypic changes than OsER1, indicating putative differentiation in their functions. The OsERL might be of essential in its function, and the proper function of all three rice ER genes might be dependent of their genetic background. Future investigations relating to these functions are key to exploiting ERfs in crop development.
Collapse
Affiliation(s)
| | | | | | | | - Ju Huang
- *Correspondence: Ju Huang, Long Wang,
| | - Long Wang
- *Correspondence: Ju Huang, Long Wang,
| |
Collapse
|
121
|
Bruder JT, Chen P, Ekberg G, Smith EC, Lazarski CA, Myers BA, Bolton J, Sedegah M, Villasante E, Richie TL, King CR, Aguiar JC, Doolan DL, Brough DE. Profiling the Targets of Protective CD8 + T Cell Responses to Infection. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:20-31. [PMID: 28948187 PMCID: PMC5602877 DOI: 10.1016/j.omtm.2017.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 11/22/2022]
Abstract
T cells are critical effectors of host immunity that target intracellular pathogens, such as the causative agents of HIV, tuberculosis, and malaria. The development of vaccines that induce effective cell-mediated immunity against such pathogens has proved challenging; for tuberculosis and malaria, many of the antigens targeted by protective T cells are not known. Here, we report a novel approach for screening large numbers of antigens as potential targets of T cells. Malaria provides an excellent model to test this antigen discovery platform because T cells are critical mediators of protection following immunization with live sporozoite vaccines and the specific antigen targets are unknown. We generated an adenovirus array by cloning 312 highly expressed pre-erythrocytic Plasmodium yoelii antigens into adenovirus vectors using high-throughput methodologies. The array was screened to identify antigen-specific CD8+ T cells induced by a live sporozoite vaccine regimen known to provide high levels of sterile protection mediated by CD8+ T cells. We identified 69 antigens that were targeted by CD8+ T cells induced by this vaccine regimen. The antigen that recalled the highest frequency of CD8+ T cells, PY02605, induced protective responses in mice, demonstrating proof of principle for this approach in identifying antigens for vaccine development.
Collapse
Affiliation(s)
- Joseph T. Bruder
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
- Corresponding author: Joseph T. Bruder, Summit Consulting, 567 Chestertown Street, Gaithersburg, MD 20878, USA.
| | - Ping Chen
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Greg Ekberg
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Emily C. Smith
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD 20817, USA
| | | | - Bennett A. Myers
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Jessica Bolton
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD 20817, USA
| | - Martha Sedegah
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Thomas L. Richie
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - C. Richter King
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| | - Joao C. Aguiar
- Malaria Department, Naval Medical Research Center (NMRC), 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
- Camris International, 3 Bethesda Metro Center, 16th Floor, Bethesda, MD 20814, USA
| | - Denise L. Doolan
- Australian Institute of Tropical Health and Medicine, James Cook University, McGregor Road, Cairns, QLD 4870, Australia
| | - Douglas E. Brough
- GenVec, Inc., 910 Clopper Road, Suite 220N, Gaithersburg, MD 20878, USA
| |
Collapse
|
122
|
Zhang R, Liu L, Yao Y, Fei F, Wang F, Yang Q, Gui Y, Wang X. High Resolution Imaging of DNA Methylation Dynamics using a Zebrafish Reporter. Sci Rep 2017; 7:5430. [PMID: 28710355 PMCID: PMC5511286 DOI: 10.1038/s41598-017-05648-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/01/2017] [Indexed: 12/17/2022] Open
Abstract
As one of the major epigenetic modifications, DNA methylation is constantly regulated during embryonic development, cell lineage commitment, and pathological processes. To facilitate real-time observation of DNA methylation, we generated a transgenic zebrafish reporter of DNA methylation (zebraRDM) via knockin of an mCherry-fused methyl-CpG binding domain (MBD) probe driven by the bactin2 promoter. The probe colocalized with heterochromatin, and its intensity was positively correlated with 5 mC immunostaining at a subcellular resolution in early embryos. Biochemical assays indicated that cells with stronger fluorescence maintained a higher level of DNA methylation, and time-lapse imaging at the blastula stage showed that the level of DNA methylation was transiently strengthened during mitosis. By crossing zebraRDM with other fluorescent transgenic lines, we demonstrate that the reporter can visually distinguish different cell lineages in organs like the heart. Our zebraRDM reporter therefore serves as a convenient and powerful tool for high-resolution investigation of methylation dynamics in live animals.
Collapse
Affiliation(s)
- Ranran Zhang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Lian Liu
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yuxiao Yao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Fei Fei
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Feng Wang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Qian Yang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yonghao Gui
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Xu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
123
|
Stark WM. Making serine integrases work for us. Curr Opin Microbiol 2017; 38:130-136. [PMID: 28599144 DOI: 10.1016/j.mib.2017.04.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 01/19/2023]
Abstract
DNA site-specific recombinases are enzymes (often associated with mobile DNA elements) that catalyse breaking and rejoining of DNA strands at specific points, thereby bringing about precise genetic rearrangements. Serine integrases are a group of recombinases derived from bacteriophages. Their unusual properties, including directionality of recombination and simple site requirements, are leading to their development as efficient, versatile tools for applications in experimental biology, biotechnology, synthetic biology and gene therapy. This article summarizes our current knowledge of serine integrase structure and mechanism, then outlines key factors that affect the performance of these phage recombination systems. Recently published studies, that have expanded the repertoire of available systems and reveal system-specific characteristics, will help us to choose the best integrases for envisaged applications.
Collapse
Affiliation(s)
- W Marshall Stark
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, Bower Building, Glasgow G12 8QQ, Scotland, United Kingdom.
| |
Collapse
|
124
|
hTERT-Immortalized Bone Mesenchymal Stromal Cells Expressing Rat Galanin via a Single Tetracycline-Inducible Lentivirus System. Stem Cells Int 2017; 2017:6082684. [PMID: 28584529 PMCID: PMC5444038 DOI: 10.1155/2017/6082684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/26/2017] [Accepted: 02/22/2017] [Indexed: 01/01/2023] Open
Abstract
The use of human telomerase reverse transcriptase-immortalized bone marrow mesenchymal stromal cells (hTERT-BMSCs) as vehicles to deliver antinociceptive galanin (GAL) molecules into pain-processing centers represents a novel cell therapy strategy for pain management. Here, an hTERT-BMSCs/Tet-on/GAL cell line was constructed using a single Tet-on-inducible lentivirus system, and subsequent experiments demonstrated that the secretion of rat GAL from hTERT-BMSCs/Tet-on/GAL was switched on and off under the control of an inducer in a dose-dependent manner. The construction of this cell line is the first promising step in the regulation of GAL secretion from hTERT-immortalized BMSCs, and the potential application of this system may provide a stem cell-based research platform for pain.
Collapse
|
125
|
Pieters T, Haenebalcke L, Bruneel K, Vandamme N, Hochepied T, van Hengel J, Wirth D, Berx G, Haigh JJ, van Roy F, Goossens S. Structure-function Studies in Mouse Embryonic Stem Cells Using Recombinase-mediated Cassette Exchange. J Vis Exp 2017. [PMID: 28518103 DOI: 10.3791/55575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Gene engineering in mouse embryos or embryonic stem cells (mESCs) allows for the study of the function of a given protein. Proteins are the workhorses of the cell and often consist of multiple functional domains, which can be influenced by posttranslational modifications. The depletion of the entire protein in conditional or constitutive knock-out (KO) mice does not take into account this functional diversity and regulation. An mESC line and a derived mouse model, in which a docking site for FLPe recombination-mediated cassette exchange (RMCE) was inserted within the ROSA26 (R26) locus, was previously reported. Here, we report on a structure-function approach that allows for molecular dissection of the different functionalities of a multidomain protein. To this end, RMCE-compatible mice must be crossed with KO mice and then RMCE-compatible KO mESCs must be isolated. Next, a panel of putative rescue constructs can be introduced into the R26 locus via RMCE targeting. The candidate rescue cDNAs can be easily inserted between RMCE sites of the targeting vector using recombination cloning. Next, KO mESCs are transfected with the targeting vector in combination with an FLPe recombinase expression plasmid. RMCE reactivates the promoter-less neomycin-resistance gene in the ROSA26 docking sites and allows for the selection of the correct targeting event. In this way, high targeting efficiencies close to 100% are obtained, allowing for insertion of multiple putative rescue constructs in a semi-high throughput manner. Finally, a multitude of R26-driven rescue constructs can be tested for their ability to rescue the phenotype that was observed in parental KO mESCs. We present a proof-of-principle structure-function study in p120 catenin (p120ctn) KO mESCs using endoderm differentiation in embryoid bodies (EBs) as the phenotypic readout. This approach enables the identification of important domains, putative downstream pathways, and disease-relevant point mutations that underlie KO phenotypes for a given protein.
Collapse
Affiliation(s)
- Tim Pieters
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Center for Medical Genetics, Ghent University Hospital; Cancer Research Institute Ghent (CRIG);
| | - Lieven Haenebalcke
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB
| | - Kenneth Bruneel
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Cancer Research Institute Ghent (CRIG)
| | - Niels Vandamme
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Cancer Research Institute Ghent (CRIG)
| | - Tino Hochepied
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB
| | - Jolanda van Hengel
- Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University
| | | | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Cancer Research Institute Ghent (CRIG)
| | - Jody J Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Alfred Centre
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Cancer Research Institute Ghent (CRIG)
| | - Steven Goossens
- Department of Biomedical Molecular Biology, Ghent University; Inflammation Research Center, VIB; Center for Medical Genetics, Ghent University Hospital; Cancer Research Institute Ghent (CRIG);
| |
Collapse
|
126
|
Germini D, Bou Saada Y, Tsfasman T, Osina K, Robin C, Lomov N, Rubtsov M, Sjakste N, Lipinski M, Vassetzky Y. A One-Step PCR-Based Assay to Evaluate the Efficiency and Precision of Genomic DNA-Editing Tools. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 5:43-50. [PMID: 28480303 PMCID: PMC5415314 DOI: 10.1016/j.omtm.2017.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/03/2017] [Indexed: 12/16/2022]
Abstract
Despite rapid progress, many problems and limitations persist and limit the applicability of gene-editing techniques. Making use of meganucleases, TALENs, or CRISPR/Cas9-based tools requires an initial step of pre-screening to determine the efficiency and specificity of the designed tools. This step remains time consuming and material consuming. Here we propose a simple, cheap, reliable, time-saving, and highly sensitive method to evaluate a given gene-editing tool based on its capacity to induce chromosomal translocations when combined with a reference engineered nuclease. In the proposed technique, designated engineered nuclease-induced translocations (ENIT), a plasmid coding for the DNA-editing tool to be tested is co-transfected into carefully chosen target cells along with that for an engineered nuclease of known specificity and efficiency. If the new enzyme efficiently cuts within the desired region, then specific chromosomal translocations will be generated between the two targeted genomic regions and be readily detectable by a one-step PCR or qPCR assay. The PCR product thus obtained can be directly sequenced, thereby determining the exact position of the double-strand breaks induced by the gene-editing tools. As a proof of concept, ENIT was successfully tested in different cell types and with different meganucleases, TALENs, and CRISPR/Cas9-based editing tools.
Collapse
Affiliation(s)
- Diego Germini
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France.,Department of Biophysics, Institute of Physics, Nanotechnology, and Telecommunications, Peter the Great St. Petersburg Polytechnic University, St. Petersburg 195251, Russia
| | - Yara Bou Saada
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France
| | - Tatiana Tsfasman
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France
| | - Kristina Osina
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,University of Latvia, 1586 Riga, Latvia
| | - Chloé Robin
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France
| | - Nikolay Lomov
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France.,M.V. Lomonosov Moscow State University, Moscow 119991, Russia
| | - Mikhail Rubtsov
- LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France.,M.V. Lomonosov Moscow State University, Moscow 119991, Russia.,Department of Biochemistry and Strategic Management Department, I.M. Sechenov First Moscow State Medical University, Moscow 119048, Russia
| | | | - Mar Lipinski
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France
| | - Yegor Vassetzky
- UMR8126, Université Paris Sud - Paris Saclay, CNRS, Institut Gustave Roussy, 94805 Villejuif, France.,LIA 1066, French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France.,M.V. Lomonosov Moscow State University, Moscow 119991, Russia
| |
Collapse
|
127
|
Wehner N, Herfert J, Dröge-Laser W, Weiste C. High-Throughput Protoplast Trans-Activation (PTA) Screening to Define Transcription Factors in Auxin-Mediated Gene Regulation. Methods Mol Biol 2017; 1569:187-202. [PMID: 28265999 DOI: 10.1007/978-1-4939-6831-2_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Genome sequencing and annotation studies clearly highlight the impact of transcriptional regulation in plants. However, functional characterization of the majority of transcriptional regulators remains elusive. Hence, high-throughput techniques are required to facilitate their molecular analysis. Here, we provide a detailed protocol to conduct a high-throughput protoplast trans-activation (PTA) screening, which enables simultaneous analysis of up to 95 individual transcription factor activities on a customizable promoter:LUCIFERASE reporter. This system is well suited to decipher complex transcriptional networks such as that triggered by the phytohormone auxin.
Collapse
Affiliation(s)
- Nora Wehner
- Pharmazeutische Biologie, Julius-von-Sachs-Institut, Julius-Maximilians-Universität Würzburg, Julius-von-Sachs Platz 2, 97082, Würzburg, Germany
| | - Jörn Herfert
- Pharmazeutische Biologie, Julius-von-Sachs-Institut, Julius-Maximilians-Universität Würzburg, Julius-von-Sachs Platz 2, 97082, Würzburg, Germany
| | - Wolfgang Dröge-Laser
- Pharmazeutische Biologie, Julius-von-Sachs-Institut, Julius-Maximilians-Universität Würzburg, Julius-von-Sachs Platz 2, 97082, Würzburg, Germany.
| | - Christoph Weiste
- Pharmazeutische Biologie, Julius-von-Sachs-Institut, Julius-Maximilians-Universität Würzburg, Julius-von-Sachs Platz 2, 97082, Würzburg, Germany
| |
Collapse
|
128
|
Dajnowicz S, Parks JM, Hu X, Gesler K, Kovalevsky AY, Mueser TC. Direct evidence that an extended hydrogen-bonding network influences activation of pyridoxal 5'-phosphate in aspartate aminotransferase. J Biol Chem 2017; 292:5970-5980. [PMID: 28232482 DOI: 10.1074/jbc.m116.774588] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/07/2017] [Indexed: 11/06/2022] Open
Abstract
Pyridoxal 5'-phosphate (PLP) is a fundamental, multifunctional enzyme cofactor used to catalyze a wide variety of chemical reactions involved in amino acid metabolism. PLP-dependent enzymes optimize specific chemical reactions by modulating the electronic states of PLP through distinct active site environments. In aspartate aminotransferase (AAT), an extended hydrogen bond network is coupled to the pyridinyl nitrogen of the PLP, influencing the electrophilicity of the cofactor. This network, which involves residues Asp-222, His-143, Thr-139, His-189, and structural waters, is located at the edge of PLP opposite the reactive Schiff base. We demonstrate that this hydrogen bond network directly influences the protonation state of the pyridine nitrogen of PLP, which affects the rates of catalysis. We analyzed perturbations caused by single- and double-mutant variants using steady-state kinetics, high resolution X-ray crystallography, and quantum chemical calculations. Protonation of the pyridinyl nitrogen to form a pyridinium cation induces electronic delocalization in the PLP, which correlates with the enhancement in catalytic rate in AAT. Thus, PLP activation is controlled by the proximity of the pyridinyl nitrogen to the hydrogen bond microenvironment. Quantum chemical calculations indicate that Asp-222, which is directly coupled to the pyridinyl nitrogen, increases the pKa of the pyridine nitrogen and stabilizes the pyridinium cation. His-143 and His-189 also increase the pKa of the pyridine nitrogen but, more significantly, influence the position of the proton that resides between Asp-222 and the pyridinyl nitrogen. These findings indicate that the second shell residues directly enhance the rate of catalysis in AAT.
Collapse
Affiliation(s)
- Steven Dajnowicz
- From the Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606.,the Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, and
| | - Jerry M Parks
- the University of Tennessee/Oak Ridge National Laboratory Center for Molecular Biophysics, Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831
| | - Xiche Hu
- From the Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606
| | - Korie Gesler
- From the Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606
| | - Andrey Y Kovalevsky
- the Biology and Soft Matter Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, and
| | - Timothy C Mueser
- From the Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606,
| |
Collapse
|
129
|
Wang X, Tang B, Ye Y, Mao Y, Lei X, Zhao G, Ding X. Bxb1 integrase serves as a highly efficient DNA recombinase in rapid metabolite pathway assembly. Acta Biochim Biophys Sin (Shanghai) 2017; 49:44-50. [PMID: 27864282 DOI: 10.1093/abbs/gmw115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/24/2016] [Indexed: 11/13/2022] Open
Abstract
Phage-encoded serine integrases are widely used in genetic engineering. They also have the potential to serve as efficient DNA assemblers, demonstrated by the method of site-specific recombination-based tandem assembly (SSRTA) that can combine biological parts into devices, pathways, and systems. Here, four serine integrases, ϕBT1, TG1, ϕRv1, and Bxb1, were investigated to ascertain their in vitro DNA assembly activities. Bxb1 integrase displayed the highest efficiency to obtain final products. Thus, we conclude that Bxb1 integrase is an excellent choice for DNA assembly in vitro Using this enzyme and its recognition sites, BioBrick standards were designed that are compatible with the SSRTA method for module addition. A rapid and efficient procedure was developed for the assembly of a multigene metabolic pathway in one step, directly from non-cutting plasmids containing the gene fragments. This technique is easy and convenient, and would be of interest to the synthetic biology community.
Collapse
Affiliation(s)
- Xianwei Wang
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Biao Tang
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- CAS Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- Institute of Quality and Standard for Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yu Ye
- Department of Microbiology and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR, China
| | - Yayi Mao
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaolai Lei
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Guoping Zhao
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- CAS Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
- Department of Microbiology and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR, China
| | - Xiaoming Ding
- Collaborative Innovation Center for Genetics and Development, State Key Laboratory of Genetic Engineering, Department of Microbiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
130
|
Dowall SD, Buttigieg KR, Findlay-Wilson SJD, Rayner E, Pearson G, Miloszewska A, Graham VA, Carroll MW, Hewson R. A Crimean-Congo hemorrhagic fever (CCHF) viral vaccine expressing nucleoprotein is immunogenic but fails to confer protection against lethal disease. Hum Vaccin Immunother 2016; 12:519-27. [PMID: 26309231 PMCID: PMC5049717 DOI: 10.1080/21645515.2015.1078045] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Crimean-Congo Hemorrhagic Fever (CCHF) is a severe tick-borne disease, endemic in many countries in Africa, the Middle East, Eastern Europe and Asia. Between 15–70% of reported cases are fatal with no approved vaccine available. In the present study, the attenuated poxvirus vector, Modified Vaccinia virus Ankara, was used to develop a recombinant candidate vaccine expressing the CCHF virus nucleoprotein. Cellular and humoral immunogenicity was confirmed in 2 mouse strains, including type I interferon receptor knockout mice, which are susceptible to CCHF disease. Despite the immune responses generated post-immunisation, the vaccine failed to protect animals from lethal disease in a challenge model.
Collapse
Affiliation(s)
- S D Dowall
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - K R Buttigieg
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | | | - E Rayner
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - G Pearson
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - A Miloszewska
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - V A Graham
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - M W Carroll
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - R Hewson
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| |
Collapse
|
131
|
Mita P, Lhakhang T, Li D, Eichinger DJ, Fenyo D, Boeke JD. Fluorescence ImmunoPrecipitation (FLIP): a Novel Assay for High-Throughput IP. Biol Proced Online 2016; 18:16. [PMID: 27528826 PMCID: PMC4983793 DOI: 10.1186/s12575-016-0046-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 07/13/2016] [Indexed: 01/27/2023] Open
Abstract
Background The immunoprecipitation (IP) assay is a valuable molecular biology tool applied across a breadth of fields. The standard assay couples IP to immunoblotting (IP/IB), a procedure severely limited as it is not easily scaled for high-throughput analysis. Results Here we describe and characterize a new methodology for fast and reliable evaluation of an immunoprecipitation reaction. FLIP (FLuorescence IP) relies on the expression of the target protein as a chromophore-tagged protein and couples IP with the measurement of fluorescent signal coating agarose beads. We show here that FLIP displays similar sensitivity to the standard IP/IB procedure but is amenable to high-throughput analysis. We applied FLIP to the screening of mouse monoclonal antibodies of unknown behavior in IP procedures. The parallel analysis of the considered antibodies using FLIP and IP/western shows good correlation between the two procedures. We also show application of FLIP using unpurified antibodies (hybridoma supernatant) and we developed a publicly available tool for the easy analysis and quantification of FLIP signals. Conclusions Altogether, our characterizations of this new methodology show that FLIP is an appealing and reliable tool for any application of high-throughput IP. Electronic supplementary material The online version of this article (doi:10.1186/s12575-016-0046-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paolo Mita
- Institute of Systems Genetics (ISG), Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center, ACLSW Room 560, 430 East 29th Street, New York, NY 10016 USA ; High Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Tenzin Lhakhang
- Center for Health Informatics and Bioinformatics, and Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center, New York, NY USA
| | - Donghui Li
- Institute of Systems Genetics (ISG), Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center, ACLSW Room 560, 430 East 29th Street, New York, NY 10016 USA ; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ; High Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | | | - David Fenyo
- Center for Health Informatics and Bioinformatics, and Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center, New York, NY USA
| | - Jef D Boeke
- Institute of Systems Genetics (ISG), Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center, ACLSW Room 560, 430 East 29th Street, New York, NY 10016 USA ; High Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
132
|
p53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PLpro via E3 ubiquitin ligase RCHY1. Proc Natl Acad Sci U S A 2016; 113:E5192-201. [PMID: 27519799 DOI: 10.1073/pnas.1603435113] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Highly pathogenic severe acute respiratory syndrome coronavirus (SARS-CoV) has developed strategies to inhibit host immune recognition. We identify cellular E3 ubiquitin ligase ring-finger and CHY zinc-finger domain-containing 1 (RCHY1) as an interacting partner of the viral SARS-unique domain (SUD) and papain-like protease (PL(pro)), and, as a consequence, the involvement of cellular p53 as antagonist of coronaviral replication. Residues 95-144 of RCHY1 and 389-652 of SUD (SUD-NM) subdomains are crucial for interaction. Association with SUD increases the stability of RCHY1 and augments RCHY1-mediated ubiquitination as well as degradation of p53. The calcium/calmodulin-dependent protein kinase II delta (CAMK2D), which normally influences RCHY1 stability by phosphorylation, also binds to SUD. In vivo phosphorylation shows that SUD does not regulate phosphorylation of RCHY1 via CAMK2D. Similarly to SUD, the PL(pro)s from SARS-CoV, MERS-CoV, and HCoV-NL63 physically interact with and stabilize RCHY1, and thus trigger degradation of endogenous p53. The SARS-CoV papain-like protease is encoded next to SUD within nonstructural protein 3. A SUD-PL(pro) fusion interacts with RCHY1 more intensively and causes stronger p53 degradation than SARS-CoV PL(pro) alone. We show that p53 inhibits replication of infectious SARS-CoV as well as of replicons and human coronavirus NL63. Hence, human coronaviruses antagonize the viral inhibitor p53 via stabilizing RCHY1 and promoting RCHY1-mediated p53 degradation. SUD functions as an enhancer to strengthen interaction between RCHY1 and nonstructural protein 3, leading to a further increase in in p53 degradation. The significance of these findings is that down-regulation of p53 as a major player in antiviral innate immunity provides a long-sought explanation for delayed activities of respective genes.
Collapse
|
133
|
Gottschamel J, Lössl A, Ruf S, Wang Y, Skaugen M, Bock R, Clarke JL. Production of dengue virus envelope protein domain III-based antigens in tobacco chloroplasts using inducible and constitutive expression systems. PLANT MOLECULAR BIOLOGY 2016; 91:497-512. [PMID: 27116001 DOI: 10.1007/s11103-016-0484-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 04/17/2016] [Indexed: 06/05/2023]
Abstract
Dengue fever is a disease in many parts of the tropics and subtropics and about half the world's population is at risk of infection according to the World Health Organization. Dengue is caused by any of the four related dengue virus serotypes DEN-1, -2, -3 and -4, which are transmitted to people by Aedes aegypti mosquitoes. Currently there is only one vaccine (Dengvaxia(®)) available (limited to a few countries) on the market since 2015 after half a century's intensive efforts. Affordable and accessible vaccines against dengue are hence still urgently needed. The dengue envelop protein domain III (EDIII), which is capable of eliciting serotype-specific neutralizing antibodies, has become the focus for subunit vaccine development. To contribute to the development of an accessible and affordable dengue vaccine, in the current study we have used plant-based vaccine production systems to generate a dengue subunit vaccine candidate in tobacco. Chloroplast genome engineering was applied to express serotype-specific recombinant EDIII proteins in tobacco chloroplasts using both constitutive and ethanol-inducible expression systems. Expression of a tetravalent antigen fusion construct combining EDIII polypeptides from all four serotypes was also attempted. Transplastomic EDIII-expressing tobacco lines were obtained and homoplasmy was verified by Southern blot analysis. Northern blot analyses showed expression of EDIII antigen-encoding genes. EDIII protein accumulation levels varied for the different recombinant EDIII proteins and the different expression systems, and reached between 0.8 and 1.6 % of total cellular protein. Our study demonstrates the suitability of the chloroplast compartment as a production site for an EDIII-based vaccine candidate against dengue fever and presents a Gateway(®) plastid transformation vector for inducible transgene expression.
Collapse
Affiliation(s)
- Johanna Gottschamel
- NIBIO-Norwegian Institute of Bioeconomy Research, P.O. Box 115, 1431, Ås, Norway
- BOKU-University of Natural Resources and Life Sciences, Gregor-Mendel-Straße 33, 1180, Vienna, Austria
| | - Andreas Lössl
- BOKU-University of Natural Resources and Life Sciences, Gregor-Mendel-Straße 33, 1180, Vienna, Austria
| | - Stephanie Ruf
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany
| | - Yanliang Wang
- NIBIO-Norwegian Institute of Bioeconomy Research, P.O. Box 115, 1431, Ås, Norway
| | | | - Ralph Bock
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam-Golm, Germany.
| | - Jihong Liu Clarke
- NIBIO-Norwegian Institute of Bioeconomy Research, P.O. Box 115, 1431, Ås, Norway.
| |
Collapse
|
134
|
Armas-Tizapantzi A, Montiel-González AM. RNAi silencing: A tool for functional genomics research on fungi. FUNGAL BIOL REV 2016. [DOI: 10.1016/j.fbr.2016.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
135
|
|
136
|
Kruis IC, Löwik DWPM, Boelens WC, van Hest JCM, Pruijn GJM. An integrated, peptide-based approach to site-specific protein immobilization for detection of biomolecular interactions. Analyst 2016; 141:5321-8. [DOI: 10.1039/c6an00154h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Site-specific immobilization of proteins on a biosensor surface, based on leucine zipper interactions.
Collapse
Affiliation(s)
- Ilmar C. Kruis
- Radboud University
- Department of Biomolecular Chemistry
- Institute for Molecules and Materials and Radboud Institute for Molecular Life Science
- Nijmegen
- The Netherlands
| | - Dennis W. P. M. Löwik
- Radboud University
- Department of Bio-organic Chemistry
- Institute for Molecules and Materials
- Nijmegen
- The Netherlands
| | - Wilbert C. Boelens
- Radboud University
- Department of Biomolecular Chemistry
- Institute for Molecules and Materials and Radboud Institute for Molecular Life Science
- Nijmegen
- The Netherlands
| | - Jan C. M. van Hest
- Radboud University
- Department of Bio-organic Chemistry
- Institute for Molecules and Materials
- Nijmegen
- The Netherlands
| | - Ger J. M. Pruijn
- Radboud University
- Department of Biomolecular Chemistry
- Institute for Molecules and Materials and Radboud Institute for Molecular Life Science
- Nijmegen
- The Netherlands
| |
Collapse
|
137
|
Abstract
Allelic exchange is an efficient method of bacterial genome engineering. This protocol describes the use of this technique to make gene knockouts and knock-ins, as well as single-nucleotide insertions, deletions and substitutions, in Pseudomonas aeruginosa. Unlike other approaches to allelic exchange, this protocol does not require heterologous recombinases to insert or excise selective markers from the target chromosome. Rather, positive and negative selections are enabled solely by suicide vector-encoded functions and host cell proteins. Here, mutant alleles, which are flanked by regions of homology to the recipient chromosome, are synthesized in vitro and then cloned into allelic exchange vectors using standard procedures. These suicide vectors are then introduced into recipient cells by conjugation. Homologous recombination then results in antibiotic-resistant single-crossover mutants in which the plasmid has integrated site-specifically into the chromosome. Subsequently, unmarked double-crossover mutants are isolated directly using sucrose-mediated counter-selection. This two-step process yields seamless mutations that are precise to a single base pair of DNA. The entire procedure requires ∼2 weeks.
Collapse
|
138
|
Recombinant Trichoderma harzianum endoglucanase I (Cel7B) is a highly acidic and promiscuous carbohydrate-active enzyme. Appl Microbiol Biotechnol 2015; 99:9591-604. [DOI: 10.1007/s00253-015-6772-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/09/2015] [Accepted: 06/12/2015] [Indexed: 12/11/2022]
|
139
|
Bagnall J, Boddington C, Boyd J, Brignall R, Rowe W, Jones NA, Schmidt L, Spiller DG, White MRH, Paszek P. Quantitative dynamic imaging of immune cell signalling using lentiviral gene transfer. Integr Biol (Camb) 2015; 7:713-25. [DOI: 10.1039/c5ib00067j] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- J. Bagnall
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - C. Boddington
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - J. Boyd
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - R. Brignall
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - W. Rowe
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - N. A. Jones
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - L. Schmidt
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - D. G. Spiller
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - M. R. H. White
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - P. Paszek
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
140
|
Dictyostelium discoideum has a highly Q/N-rich proteome and shows an unusual resilience to protein aggregation. Proc Natl Acad Sci U S A 2015; 112:E2620-9. [PMID: 25941378 DOI: 10.1073/pnas.1504459112] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many protein-misfolding diseases are caused by proteins carrying prion-like domains. These proteins show sequence similarity to yeast prion proteins, which can interconvert between an intrinsically disordered and an aggregated prion state. The natural presence of prions in yeast has provided important insight into disease mechanisms and cellular proteostasis. However, little is known about prions in other organisms, and it is not yet clear whether the findings in yeast can be generalized. Using bioinformatics tools, we show that Dictyostelium discoideum has the highest content of prion-like proteins of all organisms investigated to date, suggesting that its proteome has a high overall aggregation propensity. To study mechanisms regulating these proteins, we analyze the behavior of several well-characterized prion-like proteins, such as an expanded version of human huntingtin exon 1 (Q103) and the prion domain of the yeast prion protein Sup35 (NM), in D. discoideum. We find that these proteins remain soluble and are innocuous to D. discoideum, in contrast to other organisms, where they form cytotoxic cytosolic aggregates. However, when exposed to conditions that compromise molecular chaperones, these proteins aggregate and become cytotoxic. We show that the disaggregase Hsp101, a molecular chaperone of the Hsp100 family, dissolves heat-induced aggregates and promotes thermotolerance. Furthermore, prion-like proteins accumulate in the nucleus, where they are targeted by the ubiquitin-proteasome system. Our data suggest that D. discoideum has undergone specific adaptations that increase the proteostatic capacity of this organism and allow for an efficient regulation of its prion-like proteome.
Collapse
|
141
|
Ip LRH, Poulogiannis G, Viciano FC, Sasaki J, Kofuji S, Spanswick VJ, Hochhauser D, Hartley JA, Sasaki T, Gewinner CA. Loss of INPP4B causes a DNA repair defect through loss of BRCA1, ATM and ATR and can be targeted with PARP inhibitor treatment. Oncotarget 2015; 6:10548-62. [PMID: 25868852 PMCID: PMC4496374 DOI: 10.18632/oncotarget.3307] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 02/08/2015] [Indexed: 12/18/2022] Open
Abstract
Treatment options for ovarian cancer patients remain limited and overall survival is less than 50% despite recent clinical advances. The lipid phosphatase inositol polyphosphate 4-phosphatase type II (INPP4B) has been described as a tumor suppressor in the PI3K/Akt pathway with loss of expression found most pronounced in breast, ovarian cancer and melanoma. Using microarray technology we identified a DNA repair defect in INPP4B-deficient cells, which we further characterized by comet assays and quantification of γH2AX, RAD51 and 53BP1 foci formation. INPP4B loss resulted in significantly increased sensitivity towards PARP inhibition, comparable to loss of BRCA1 in two- and three-dimensional in vitro models, as well as in in vivo xenograft models. Mechanistically, we discovered that INPP4B forms a protein complex with the key players of DNA repair, ATR and BRCA1, in GST pulldown and 293T overexpression assays, and INPP4B loss affects BRCA1, ATM and ATR protein stability resulting in the observed DNA repair defect. Given that INPP4B loss has been found in 40% of ovarian cancer patients, this study provides the rationale for establishing INPP4B as a biomarker of PARP inhibitor response, and consequently offers novel therapeutic options for a significant subset of patients. Loss of the tumor suppressor inositol polyphosphate 4-phosphatase type II (INPP4B) results in a DNA repair defect due to concomitant loss of BRCA1, ATR and ATM and can be therapeutically targeted with PARP inhibitors.
Collapse
Affiliation(s)
- Laura R H Ip
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK
| | - George Poulogiannis
- The Institute of Cancer Research, Signalling and Cancer Metabolism, London, UK
| | - Felipe Cia Viciano
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK
- Faculty of Infectious and Tropical Diseases, Immunology and Infection Department, London School of Hygiene & Tropical Diseases, London, UK
| | - Junko Sasaki
- Department of Medical Biology, Akita University School of Medicine, Akita, Japan
| | - Satoshi Kofuji
- Department of Medical Biology, Akita University School of Medicine, Akita, Japan
| | - Victoria J Spanswick
- Cancer Research UK Drug-DNA Interaction Research Group, UCL Cancer Institute, University College London, London, UK
| | - Daniel Hochhauser
- Cancer Research UK Drug-DNA Interaction Research Group, UCL Cancer Institute, University College London, London, UK
| | - John A Hartley
- Cancer Research UK Drug-DNA Interaction Research Group, UCL Cancer Institute, University College London, London, UK
| | - Takehiko Sasaki
- Department of Medical Biology, Akita University School of Medicine, Akita, Japan
| | - Christina A Gewinner
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
142
|
Wille T, Barlag B, Jakovljevic V, Hensel M, Sourjik V, Gerlach RG. A gateway-based system for fast evaluation of protein-protein interactions in bacteria. PLoS One 2015; 10:e0123646. [PMID: 25856398 PMCID: PMC4391838 DOI: 10.1371/journal.pone.0123646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/26/2015] [Indexed: 11/18/2022] Open
Abstract
Protein-protein interactions are important layers of regulation in all kingdoms of life. Identification and characterization of these interactions is one challenging task of the post-genomic era and crucial for understanding of molecular processes within a cell. Several methods have been successfully employed during the past decades to identify protein-protein interactions in bacteria, but most of them include tedious and time-consuming manipulations of DNA. In contrast, the MultiSite Gateway system is a fast tool for transfer of multiple DNA fragments between plasmids enabling simultaneous and site directed cloning of up to four fragments into one construct. Here we developed a new set of Gateway vectors including custom made entry vectors and modular Destination vectors for studying protein-protein interactions via Fluorescence Resonance Energy Transfer (FRET), Bacterial two Hybrid (B2H) and split Gaussia luciferase (Gluc), as well as for fusions with SNAP-tag and HaloTag for dual-color super-resolution microscopy. As proof of principle, we characterized the interaction between the Salmonella effector SipA and its chaperone InvB via split Gluc and B2H approach. The suitability for FRET analysis as well as functionality of fusions with SNAP- and HaloTag could be demonstrated by studying the transient interaction between chemotaxis response regulator CheY and its phosphatase CheZ.
Collapse
Affiliation(s)
- Thorsten Wille
- Junior Research Group 3, Robert Koch-Institute, Wernigerode Branch, Wernigerode, Germany
| | - Britta Barlag
- Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Vladimir Jakovljevic
- Center for Molecular Biology at the University of Heidelberg (ZMBH), DKFZ (German Cancer Research Center) -ZMBH Alliance, Heidelberg, Germany
| | - Michael Hensel
- Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Victor Sourjik
- Center for Molecular Biology at the University of Heidelberg (ZMBH), DKFZ (German Cancer Research Center) -ZMBH Alliance, Heidelberg, Germany
- Max Planck Institute for Terrestrial Microbiology & LOEWE (state offensive for the development of scientific and economic excellence) Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Roman G. Gerlach
- Junior Research Group 3, Robert Koch-Institute, Wernigerode Branch, Wernigerode, Germany
- * E-mail:
| |
Collapse
|
143
|
Albers J, Danzer C, Rechsteiner M, Lehmann H, Brandt LP, Hejhal T, Catalano A, Busenhart P, Gonçalves AF, Brandt S, Bode PK, Bode-Lesniewska B, Wild PJ, Frew IJ. A versatile modular vector system for rapid combinatorial mammalian genetics. J Clin Invest 2015; 125:1603-19. [PMID: 25751063 PMCID: PMC4396471 DOI: 10.1172/jci79743] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 01/20/2015] [Indexed: 01/29/2023] Open
Abstract
Here, we describe the multiple lentiviral expression (MuLE) system that allows multiple genetic alterations to be introduced simultaneously into mammalian cells. We created a toolbox of MuLE vectors that constitute a flexible, modular system for the rapid engineering of complex polycistronic lentiviruses, allowing combinatorial gene overexpression, gene knockdown, Cre-mediated gene deletion, or CRISPR/Cas9-mediated (where CRISPR indicates clustered regularly interspaced short palindromic repeats) gene mutation, together with expression of fluorescent or enzymatic reporters for cellular assays and animal imaging. Examples of tumor engineering were used to illustrate the speed and versatility of performing combinatorial genetics using the MuLE system. By transducing cultured primary mouse cells with single MuLE lentiviruses, we engineered tumors containing up to 5 different genetic alterations, identified genetic dependencies of molecularly defined tumors, conducted genetic interaction screens, and induced the simultaneous CRISPR/Cas9-mediated knockout of 3 tumor-suppressor genes. Intramuscular injection of MuLE viruses expressing oncogenic H-RasG12V together with combinations of knockdowns of the tumor suppressors cyclin-dependent kinase inhibitor 2A (Cdkn2a), transformation-related protein 53 (Trp53), and phosphatase and tensin homolog (Pten) allowed the generation of 3 murine sarcoma models, demonstrating that genetically defined autochthonous tumors can be rapidly generated and quantitatively monitored via direct injection of polycistronic MuLE lentiviruses into mouse tissues. Together, our results demonstrate that the MuLE system provides genetic power for the systematic investigation of the molecular mechanisms that underlie human diseases.
Collapse
|
144
|
In-Frame and Unmarked Gene Deletions in Burkholderia cenocepacia via an Allelic Exchange System Compatible with Gateway Technology. Appl Environ Microbiol 2015; 81:3623-30. [PMID: 25795676 DOI: 10.1128/aem.03909-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 03/11/2015] [Indexed: 02/04/2023] Open
Abstract
Burkholderia cenocepacia is an emerging opportunistic pathogen causing life-threatening infections in immunocompromised individuals and in patients with cystic fibrosis, which are often difficult, if not impossible, to treat. Understanding the genetic basis of virulence in this emerging pathogen is important for the development of novel treatment regimes. Generation of deletion mutations in genes predicted to encode virulence determinants is fundamental to investigating the mechanisms of pathogenesis. However, there is a lack of appropriate selectable and counterselectable markers for use in B. cenocepacia, making its genetic manipulation problematic. Here we describe a Gateway-compatible allelic exchange system based on the counterselectable pheS gene and the I-SceI homing endonuclease. This system provides efficiency in cloning homology regions of target genes and allows the generation of precise and unmarked gene deletions in B. cenocepacia. As a proof of concept, we demonstrate its utility by deleting the Bcam1349 gene, encoding a cyclic di-GMP (c-di-GMP)-responsive regulator protein important for biofilm formation.
Collapse
|
145
|
LaCava J, Molloy KR, Taylor MS, Domanski M, Chait BT, Rout MP. Affinity proteomics to study endogenous protein complexes: pointers, pitfalls, preferences and perspectives. Biotechniques 2015; 58:103-19. [PMID: 25757543 PMCID: PMC4465938 DOI: 10.2144/000114262] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/17/2015] [Indexed: 01/13/2023] Open
Abstract
Dissecting and studying cellular systems requires the ability to specifically isolate distinct proteins along with the co-assembled constituents of their associated complexes. Affinity capture techniques leverage high affinity, high specificity reagents to target and capture proteins of interest along with specifically associated proteins from cell extracts. Affinity capture coupled to mass spectrometry (MS)-based proteomic analyses has enabled the isolation and characterization of a wide range of endogenous protein complexes. Here, we outline effective procedures for the affinity capture of protein complexes, highlighting best practices and common pitfalls.
Collapse
Affiliation(s)
- John LaCava
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York
- Institute for Systems Genetics, New York University School of Medicine, New York, NY
| | - Kelly R. Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY
| | - Martin S. Taylor
- High Throughput Biology Center and Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michal Domanski
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York
- Centre for mRNP Biogenesis and Metabolism, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Brian T. Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY
| | - Michael P. Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York
| |
Collapse
|
146
|
Bruckbauer ST, Kvitko BH, Karkhoff-Schweizer RR, Schweizer HP. Tn5/7-lux: a versatile tool for the identification and capture of promoters in gram-negative bacteria. BMC Microbiol 2015; 15:17. [PMID: 25648327 PMCID: PMC4328036 DOI: 10.1186/s12866-015-0354-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The combination of imaging technologies and luciferase-based bioluminescent bacterial reporter strains provide a sensitive and simple non-invasive detection method (photonic bioimaging) for the study of diverse biological processes, as well as efficacy of therapeutic interventions, in live animal models of disease. The engineering of bioluminescent bacteria required for photonic bioimaging is frequently hampered by lack of promoters suitable for strong, yet stable luciferase gene expression. RESULTS We devised a novel method for identification of constitutive native promoters in Gram-negative bacteria. The method is based on a Tn5/7 transposon that exploits the unique features of Tn5 (random transposition) and Tn7 (site-specific transposition). The transposons are designed such that Tn5 transposition will allow insertion of a promoter-less bacterial luxCDABE operon downstream of a bacterial gene promoter. Cloning of DNA fragments from luminescent isolates results in a plasmid that replicates in pir (+) hosts. Sequencing of the lux-chromosomal DNA junctions on the plasmid reveals transposon insertion sites within genes or operons. The plasmid is also a mini-Tn7-lux delivery vector that can be used to introduce the promoter-lux operon fusion into other derivatives of the bacterium of interest in an isogenic fashion. Alternatively, promoter-containing sequences can be PCR-amplified from plasmid or chromosomal DNA and cloned into a series of accompanying mini-Tn7-lux vectors. The mini-Tn5/7-lux and mini-Tn7-lux vectors are equipped with diverse selection markers and thus applicable in numerous Gram-negative bacteria. Various mini-Tn5/7-lux vectors were successfully tested for transposition and promoter identification by imaging in Acinetobacter baumannii, Escherichia coli, and Burkholderia pseudomallei. Strong promoters were captured for lux expression in E. coli and A. baumannii. Some mini-Tn7-lux vectors are also equipped with attB sites for swapping of the lux operon with other reporter genes using Gateway technology. CONCLUSIONS Although mini-Tn5-lux and mini-Tn7-lux elements have previously been developed and used for bacterial promoter identification and chromosomal insertion of promoter-lux gene fusions, respectively, the newly developed mini-Tn5/7-lux and accompanying accessory plasmids streamline and accelerate the promoter discovery and bioluminescent strain engineering processes. Availability of vectors with diverse selection markers greatly extend the host-range of promoter probe and lux gene fusion vectors.
Collapse
Affiliation(s)
- Steven T Bruckbauer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Present Address: Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, 53706, WI, USA.
| | - Brian H Kvitko
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Present Address: MSU-DOE Plant Research Laboratory, Michigan State University, East Lansing, 48824, MI, USA.
| | - RoxAnn R Karkhoff-Schweizer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA.
| | - Herbert P Schweizer
- Department of Microbiology, Immunology and Pathology, and Rocky Mountain Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research, Colorado State University, Fort Collins, 80523, CO, USA. .,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Emerging Pathogens Institute, PO Box 100266, Gainesville, 32610-0266, FL, USA.
| |
Collapse
|
147
|
Calçada EO, Korsak M, Kozyreva T. Recombinant Intrinsically Disordered Proteins for NMR: Tips and Tricks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 870:187-213. [PMID: 26387103 DOI: 10.1007/978-3-319-20164-1_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The growing recognition of the several roles that intrinsically disordered proteins play in biology places an increasing importance on protein sample availability to allow the characterization of their structural and dynamic properties. The sample preparation is therefore the limiting step to allow any biophysical method being able to characterize the properties of an intrinsically disordered protein and to clarify the links between these properties and the associated biological functions. An increasing array of tools has been recruited to help prepare and characterize the structural and dynamic properties of disordered proteins. This chapter describes their sample preparation, covering the most common drawbacks/barriers usually found working in the laboratory bench. We want this chapter to be the bedside book of any scientist interested in preparing intrinsically disordered protein samples for further biophysical analysis.
Collapse
Affiliation(s)
- Eduardo O Calçada
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy.
| | - Magdalena Korsak
- Giotto Biotech, Via Madonna del Piano 6, 50019, Sesto Fiorentino, Italy.
| | - Tatiana Kozyreva
- Giotto Biotech, Via Madonna del Piano 6, 50019, Sesto Fiorentino, Italy.
| |
Collapse
|
148
|
Lehtonen SI, Taskinen B, Ojala E, Kukkurainen S, Rahikainen R, Riihimaki TA, Laitinen OH, Kulomaa MS, Hytonen VP. Efficient preparation of shuffled DNA libraries through recombination (Gateway) cloning. Protein Eng Des Sel 2014; 28:23-8. [DOI: 10.1093/protein/gzu050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
149
|
Recombinant expressed vector pET32a (+) S constructed by ligation independent cloning. Molecules 2014; 19:16179-89. [PMID: 25310147 PMCID: PMC6271046 DOI: 10.3390/molecules191016179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/09/2014] [Accepted: 09/09/2014] [Indexed: 11/17/2022] Open
Abstract
The aim of this work was to develop a new method for constructing vectors, named ligation-independent cloning (LIC) method. We constructed the S label expression vector and recombinant pET32a (+) S-phoN2 by LIC. The recombinant proteins were expressed in E. coli at a high level, and then the specificity of the recombinant proteins was identified by western blot. The target band was detected by S monoclonal antibody and Apyrase polyclonal antibodies but not Trx monoclonal antibody and HIS monoclonal antibody. Finally, we obtained protein Apyrase in E. coli (BL21), with a protein-only expression S tag. Collectively, our results demonstrated that LIC is effective for the construction of new vectors and recombinant plasmids. Free from the limitations of restriction enzyme sites and with a higher positive rate, LIC processes should find broad applications in molecular biology research.
Collapse
|
150
|
Abstract
ABSTRACT
Since the discovery of restriction enzymes and the generation of the first recombinant DNA molecule over 40 years ago, molecular biology has evolved into a multidisciplinary field that has democratized the conversion of a digitized DNA sequence stored in a computer into its biological counterpart, usually as a plasmid, stored in a living cell. In this article, we summarize the most relevant tools that allow the swift assembly of DNA sequences into useful plasmids for biotechnological purposes. We cover the main components and stages in a typical DNA assembly workflow, namely
in silico
design,
de novo
gene synthesis, and
in vitro
and
in vivo
sequence assembly methodologies.
Collapse
|