plant genome editing with CRISPR systems, 1st ed , yiping qi, 2019 41

359 26 0
 plant genome editing with CRISPR systems, 1st ed , yiping qi, 2019   41

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Methods in Molecular Biology 1917 Yiping Qi Editor Plant Genome Editing with CRISPR Systems Methods and Protocols METHODS IN MOLECULAR BIOLOGY Series Editor John M Walker School of Life and Medical Sciences University of Hertfordshire Hatfield, Hertfordshire, AL10 9AB, UK For further volumes: http://www.springer.com/series/7651 Plant Genome Editing with CRISPR Systems Methods and Protocols Edited by Yiping Qi Department of Plant Science and Landscape Architecture, University of Maryland College Park, College Park, MD, USA Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, USA Editor Yiping Qi Department of Plant Science and Landscape Architecture University of Maryland College Park College Park, MD, USA Institute for Bioscience and Biotechnology Research University of Maryland Rockville, MD, USA ISSN 1064-3745 ISSN 1940-6029 (electronic) Methods in Molecular Biology ISBN 978-1-4939-8990-4 ISBN 978-1-4939-8991-1 (eBook) https://doi.org/10.1007/978-1-4939-8991-1 Library of Congress Control Number: 2018965601 © Springer Science+Business Media, LLC, part of Springer Nature 2019 This work is subject to copyright All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed The use of general descriptive names, registered names, trademarks, service marks, etc in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication Neither the publisher nor the authors or the editors give a warranty, express or implied, with respect to the material contained herein or for any errors or omissions that may have been made The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer Nature The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A Preface The world has witnessed a great period of food crop productivity growth in the past 50 years Notably, the introduction of crop genetic improvement technologies into the developing world has resulted in drastic yield increases for major staple crops such as wheat and rice This achievement is remembered as the Green Revolution (1966–1985) Afterwards, recombinant DNA-based biotechnology contributed to the development of highly efficient genetically modified (GM) crops, thanks to pioneers like Mary-Dell Chilton who co-developed Agrobacterium-mediated plant transformation technology However, GM crops are expensive to develop, and they also face public acceptance problems in many countries Meanwhile, conventional breeding cannot keep pace with global population growth and climate change For example, the current rate of annual yield increases for four major crops (wheat, rice, maize, and soybean) must be doubled to meet the future demand in 2050 All these challenges call for the development of new breeding technologies that can potentially revolutionize agriculture Genome editing is one such technology Genome editing enables rewriting the DNA sequence in a genome, which in most cases relies on the ability to make DNA double strand breaks (DSBs) in a sequence-specific manner Sequence-specific nucleases (SSNs) are molecular scissors that are engineered to make targeted DNA DSBs SSNs such as zinc finger nuclease (ZFN), transcription activatorlike effector nuclease (TALEN), and CRISPR (clustered regularly interspaced short palindromic repeats)-Cas systems have been successfully applied in many plant species to achieve efficient genome editing Because CRISPR-Cas is guided by a custom-designed guide RNA to recognize and cleave the target DNA, this mechanism drastically simplifies the engineering process of a customized SSN, making CRISPR-Cas the top choice for plant genome editing Developed in 2012 and applied to eukaryotic cells in 2013, CRISPR-Cas genome editing technology has since been revolutionizing plant biology It boosts reverse genetics research in non-model plants and represents an efficient breeding technology for crop improvement In recent years, the number of peer-reviewed papers utilizing CRISPR in plants has skyrocketed Yet, it can be difficult and confusing for new users to choose a CRISPR system in order to achieve a specific genome editing outcome in a plant of interest To help readers who are interested in learning and using CRISPR systems in plants, this book series provides comprehensive coverage of CRISPR systems and applications in different plant species The book starts with a review on plant DNA repair and genome editing by Qiudeng Que, Mary-Dell Chilton, and their colleagues (Chapter 1) The remaining chapters document methods and protocols on analysis of CRISPR-induced mutations (Chapters and 3), multiplexed CRISPR-Cas9 systems (Chapters 4–7), CRISPR-Cas9 editing in monocots (e.g., rice and maize; Chapters 8–10), CRISPR-Cas9 editing in dicots (e.g., Arabidopsis, Brassica oleracea, tomato, potato, carrot, soybean, and citrus; Chapters 11–17), CRISPRCas12a (Cpf1) editing systems (Chapters 18–20), precise gene editing (e.g., gene replacement and base editing; Chapters 21 and 22), and non-Agrobacterium-based CRISPR delivery systems (e.g., virus delivery, ribonucleoprotein (RNP) delivery to calli or protoplasts, and automated protoplast transformation; Chapters 23–26) v vi Preface I thank Aimee Malzahn for making the artistic cover picture and all the authors for making great contributions to this book I would like to give special thanks to my wife, Hong Chen, for her generous support to my work and also to my former postdoc mentor, Daniel Voytas, for his introduction of my career into plant genome editing, an important and exciting field College Park, MD, USA Yiping Qi Contents Preface Contributors PART I REVIEW ON PLANT DNA REPAIR AND GENOME EDITING Plant DNA Repair Pathways and Their Applications in Genome Engineering Qiudeng Que, Zhongying Chen, Tim Kelliher, David Skibbe, Shujie Dong, and Mary-Dell Chilton PART II 27 33 MULTIPLEXED CRISPR-CAS9 SYSTEMS Creating Large Chromosomal Deletions in Rice Using CRISPR/Cas9 Riqing Li, Si Nian Char, and Bing Yang A Multiplexed CRISPR/Cas9 Editing System Based on the Endogenous tRNA Processing Kabin Xie and Yinong Yang A Single Transcript CRISPR-Cas9 System for Multiplex Genome Editing in Plants Xu Tang, Zhaohui Zhong, Qiurong Ren, Binglin Liu, and Yong Zhang CRISPR-Act2.0: An Improved Multiplexed System for Plant Transcriptional Activation Aimee Malzahn, Yong Zhang, and Yiping Qi PART IV CRISPR DESIGN AND MUTATION ANALYSIS Rapid Screening of CRISPR/Cas9-Induced Mutants Using the ACT-PCR Method Chun Wang and Kejian Wang Decoding Sanger Sequencing Chromatograms from CRISPR-Induced Mutations Xianrong Xie, Xingliang Ma, and Yao-Guang Liu PART III v xi 47 63 75 83 CRISPR-CAS9 EDITING IN MONOCOTS Generating Photoperiod-Sensitive Genic Male Sterile Rice Lines with CRISPR/Cas9 97 Weihang Gu, Dabing Zhang, Yiping Qi, and Zheng Yuan Knocking Out MicroRNA Genes in Rice with CRISPR-Cas9 109 Jianping Zhou, Zhaohui Zhong, Hongqiao Chen, Qian Li, Xuelian Zheng, Yiping Qi, and Yong Zhang vii viii 10 Contents An Agrobacterium-Mediated CRISPR/Cas9 Platform for Genome Editing in Maize 121 Keunsub Lee, Huilan Zhu, Bing Yang, and Kan Wang PART V 11 12 13 14 15 16 17 Fluorescence Marker-Assisted Isolation of Cas9-Free and CRISPR-Edited Arabidopsis Plants Hanchuanzhi Yu and Yunde Zhao Creating Targeted Gene Knockouts in Brassica oleracea Using CRISPR/Cas9 Tom Lawrenson, Penny Hundleby, and Wendy Harwood Application of CRISPR/Cas9-Mediated Gene Editing in Tomato Nathan T Reem and Joyce Van Eck Genome Editing in Potato with CRISPR/Cas9 Satya Swathi Nadakuduti, Colby G Starker, Daniel F Voytas, C Robin Buell, and David S Douches Visual Assay for Gene Editing Using a CRISPR/Cas9 System in Carrot Cells Magdalena Klimek-Chodacka, Tomasz Oleszkiewicz, and Rafal Baranski Genome Editing in Soybean with CRISPR/Cas9 Junqi Liu, Samatha Gunapati, Nicole T Mihelich, Adrian O Stec, Jean-Michel Michno, and Robert M Stupar Genome Editing in Citrus Tree with CRISPR/Cas9 Hongge Jia, Xiuping Zou, Vladimir Orbovic, and Nian Wang PART VI 18 19 20 22 147 155 171 183 203 217 235 CRISPR-CAS12A EDITING SYSTEMS Plant Gene Knockout and Knockdown by CRISPR-Cpf1 (Cas12a) Systems 245 Yingxiao Zhang, Yong Zhang, and Yiping Qi Editing a Stomatal Developmental Gene in Rice with CRISPR/Cpf1 257 Xiaojia Yin, Abhishek Anand, Paul Quick, and Anindya Bandyopadhyay Targeted Mutagenesis Using FnCpf1 in Tobacco 269 Akira Endo and Seiichi Toki PART VII 21 CRISPR-CAS9 EDITING IN DICOTS PRECISE GENE EDITING Gene Replacement by Intron Targeting with CRISPR-Cas9 285 Jun Li, Xiangbing Meng, Jiayang Li, and Caixia Gao Targeted Base Editing with CRISPR-Deaminase in Tomato 297 Zenpei Shimatani, Tohru Ariizumi, Ushio Fujikura, Akihiko Kondo, Hiroshi Ezura, and Keiji Nishida Contents PART VIII 23 24 25 26 ix NON-AGROBACTERIUM BASED CRISPR DELIVERY SYSTEMS Virus-Mediated Genome Editing in Plants Using the CRISPR/Cas9 System Ahmed Mahas, Zahir Ali, Manal Tashkandi, and Magdy M Mahfouz Biolistic Delivery of CRISPR/Cas9 with Ribonucleoprotein Complex in Wheat Zhen Liang, Kunling Chen, and Caixia Gao DNA-Free Genome Editing via Ribonucleoprotein (RNP) Delivery of CRISPR/Cas in Lettuce Jongjin Park, Sunmee Choi, Slki Park, Jiyoung Yoon, Aiden Y Park, and Sunghwa Choe An Automated Protoplast Transformation System Scott C Lenaghan and C Neal Stewart Jr Index 311 327 337 355 365 Contributors ZAHIR ALI  Laboratory for Genome Engineering, Division of Environmental and Biological Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia ABHISHEK ANAND  International Rice Research Institute, Manila, Philippines TOHRU ARIIZUMI  Faculty of Life and Environmental Sciences, Gene Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan ANINDYA BANDYOPADHYAY  International Rice Research Institute, Manila, Philippines; Syngenta Beijing Innovation Center, Beijing, China RAFAL BARANSKI  Faculty of Biotechnology and Horticulture, Institute of Plant Biology and Biotechnology, University of Agriculture in Krakow, Krakow, Poland C ROBIN BUELL  Department of Plant Biology, Michigan State University, East Lansing, MI, USA; Plant Resilience Institute, Michigan State University, East Lansing, MI, USA; Michigan State University AgBioResearch, Michigan State University, East Lansing, MI, USA SI NIAN CHAR  Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA HONGQIAO CHEN  Department of Biotechnology, School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China KUNLING CHEN  State Key Laboratory of Plant Cell and Chromosome Engineering, Center for Genome Editing, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China ZHONGYING CHEN  Seeds Research, Syngenta Crop Protection, LLC, Research Triangle Park, NC, USA MARY-DELL CHILTON  Seeds Research, Syngenta Crop Protection, LLC, Research Triangle Park, NC, USA SUNGHWA CHOE  Naturegenic Inc., West Lafayette, IN, USA; G+FLAS Life Sciences, Seoul, South Korea; School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea SUNMEE CHOI  G+FLAS Life Sciences, Seoul, South Korea SHUJIE DONG  Seeds Research, Syngenta Crop Protection, LLC, Research Triangle Park, NC, USA DAVID S DOUCHES  Department of Plant, Soil and Microbial Sciences, Michigan State University, East Lansing, MI, USA; Michigan State University AgBioResearch, Michigan State University, East Lansing, MI, USA AKIRA ENDO  Plant Genome Engineering Research Unit, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan HIROSHI EZURA  Faculty of Life and Environmental Sciences, Gene Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan USHIO FUJIKURA  Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Hyogo, Japan xi 352 Jongjin Park et al Fig Time course morphology of regenerating protoplasts (a) Five-day-old protoplasts after transfection with RNPs; the protoplasts are doubled at days (b) The protoplasts form colonies at seventh day (c) Microcalli (d) Calli (e) Calli turn green after weeks under light (f) Plantlets with shoots regenerated Bars ¼ 100 μm and 0.5 cm 18 After weeks, transfer low-melting agar with microcalli to Shoot Induction Media (SIM) (see Fig 3d) SIM 1L MS powder 4.4 g Sucrose 30 g 0.1 mg NAA 100 μL (1 mg/mL stock) 0.5 mg BAP 500 μL (0.1 mg/mL stock) Plant agar 6g Ribonucleoprotein Delivery of CRISPR/Cas 353 19 After weeks, transfer calli from SIM to MS media and keep under light (see Fig 3e) 20 When shoots emerge, transfer the tiny plantlets into MS media (see Fig 3f) 21 Transfer rooted plants to soil 22 Screen edited events among regenerated plants (see Note 4) Notes An alternative method for preparing transcription template for SpCas9 sgRNA: A plasmid carrying T7 promoter and guide RNA scaffold is constructed Only a target 20 bp doublestranded oligonucleotide is cloned into the middle of two BsaI sites (A#TAGGTGAGACCGCAGGTCTCG#GTTTT) placed between T7 promoter and guide RNA scaffold by two BsaI type IIS restriction enzyme from Golden Gate cloning method (see Fig 1a) A forward single oligonucleotide should embody 50 -TAGG-30 overhang in front of the target 20 nt, while a reverse single oligonucleotide gets initiated with 50 -CAAA-30 in front of the reverse target 20 nt Both one picomole of forward and reverse single oligonucleotides are mixed in 45 μL distilled water, which is transferred into 0.2 mL PCR tube, and anneal at 95  C for and 55  C for 10 by a thermocycler, then place annealed oligonucleotides on ice As a result, the dimerized oligonucleotides are employed to clone into a linear plasmid with two flanking sequences, 50 -CCTA-30 and 50 -GTTT-30 The completed construct is used to synthesize sgRNAs as templates An alternative method to prepare a transcription template for FnCpf1 crRNA: Synthesize two 63 nt single-stranded oligonucleotides, which compose of nt overhang in front of T7 promoter, 19 nt T7 promoter, and 20 nt target spacer sequence Both 10 μL of 200 nmol of forward and reverse single oligonucleotides are mixed, and the 20 μL mixture is transferred into a 0.2 mL PCR tube and annealed at 95  C for and 55  C for 10 by a thermocycler, then place annealed dsODN on ice It is strongly recommended to wear gloves and use nucleasefree tubes and reagents to avoid RNase contamination Reactions are typically 20 μL but can be scaled up as needed Reactions should be assembled in nuclease-free micro centrifuge tubes or PCR strip tubes In the earlier chapters of this book, different genotyping methods are described for screening CRISPR-induced mutations Readers can refer to these chapters for details 354 Jongjin Park et al References Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E (2012) A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity Science 337 (6096):816–821 Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri S, Yang L, Church GM (2013) CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering Nat Biotechnol 31(9):833–838 Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N, Ishitani R, Zhang F, Nureki O (2014) Crystal structure of Cas9 in complex with guide RNA and target DNA Cell 156(5):935–949 Ma E, Harrington LB, O’Connell MR, Zhou K, Doudna JA (2015) Single-stranded DNA cleavage by divergent CRISPR-Cas9 enzymes Mol Cell 60(3):398–407 Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, Volz SE, Joung J, van der Oost J, Regev A et al (2015) Cpf1 is a single RNA-guided endonuclease of a class CRISPR-Cas system Cell 163(3):759–771 Fagerlund RD, Staals RH, Fineran PC (2015) The Cpf1 CRISPR-Cas protein expands genome-editing tools Genome Biol 16:251 Ikeuchi M, Ogawa Y, Iwase A, Sugimoto K (2016) Plant regeneration: cellular origins and molecular mechanisms Development 143 (9):1442–1451 Woo JW, Kim J, Kwon SI, Corvalan C, Cho SW, Kim H, Kim SG, Kim ST, Choe S, Kim JS (2015) DNA-free genome editing in plants with preassembled CRISPR-Cas9 ribonucleoproteins Nat Biotechnol 33(11):1162–1164 Kim H, Kim ST, Ryu J, Kang BC, Kim JS, Kim SG (2017) CRISPR/Cpf1-mediated DNAfree plant genome editing Nat Commun 8:14406 10 Anders C, Jinek M (2014) In vitro enzymology of Cas9 Methods Enzymol 546:1–20 Chapter 26 An Automated Protoplast Transformation System Scott C Lenaghan and C Neal Stewart Jr Abstract Efficient plant protoplast production from cell suspension cultures, leaf, and stem tissue allows for singlecell plant biology Since protoplasts not have cell walls, they can be readily transformed to enable rapid assessment of regulatory elements, synthetic constructs, gene expression, and more recently genomeediting tools and approaches Historically, enzymatic cell wall digestion has been both expensive and laborious Protoplast production, transformation, and analysis of fluorescence have recently been automated using an integrated robotic system Here we describe its use for bulk protoplast isolation, counting, transformation, and analysis at very low cost for high-throughput experiments Key words Tobacco, Protoplasts, Transformation, Enzymatic digestion, High-throughput screening, Automation, Robotics Introduction Significant effort has been placed on generating crops with advantageous traits, including disease resistance [1], herbicide resistance [2], drought [3, 4] and salt tolerance [5], increased biomass [6], and altered cell wall structure [7] With the advent of a new generation of tools for molecular breeding, including CRISPR-Cas9 and TALENs [8], along with more traditional gene silencing tools, such as dsRNA [9], miRNA [10], and siRNA [11], a bottleneck has been created whereby more plants can be generated than could possibly be screened In particular, the low cost of generating guide RNA targets (gRNAs) for CRISPR-Cas9 means that researchers can generate hundreds to thousands of constructs, and thus could theoretically rapidly generate similar number of transgenic plants Unfortunately, while the technical and cost barriers for generation of the constructs are decreasing, the costs and space requirements to screen thousands of plants is extremely high Further, many of these targets will lead to undesirable effects that ideally would be identified at an earlier stage As such, plant protoplasts have emerged as plants’ answer to single-cell biology Protoplast Yiping Qi (ed.), Plant Genome Editing with CRISPR Systems: Methods and Protocols, Methods in Molecular Biology, vol 1917, https://doi.org/10.1007/978-1-4939-8991-1_26, © Springer Science+Business Media, LLC, part of Springer Nature 2019 355 356 Scott C Lenaghan and C Neal Stewart Jr platforms are especially useful for high-throughput gene expression assays, for genome-editing, gene silencing, and a variety of other molecular breeding approaches Protoplasts have several advantages compared to the use of intact plant tissue or cell suspension cultures The primary advantage is the lack of a cell wall, which is a significant barrier that leads to the generally poor transformation efficiency of plant cells [12] In addition, protoplasts represent a true single-cell culture, as opposed to plant cell suspension cultures and plant tissue, which are multicellular The final advantage of protoplasts is the potential to extract protoplasts from nearly any organs and tissues from whole plants, thus representing the developmental and spatial features inherent in those organs and tissues This enables the identification of tissue-specific expression, the assessment of functional chloroplasts, and the ability to look at gene expression from different tissues In addition, protoplasts can also be isolated from homogeneous cell suspension cultures, which are primarily derived from callus [13] Based on the utility of protoplasts for early screening of molecular targets, a protocol was developed for highthroughput, automated protoplast isolation, transformation, and screening [14] In this work the widely used tobacco (Nicotiana tabacum L.) ‘Bright Yellow’ (BY-2) suspension culture was used as a model to demonstrate the approach In this chapter, we will describe a general protocol for automated protoplast isolation, PEG-mediated transformation, and screening using BY-2 as the model system While any automated protocol will be dependent on the equipment available to carry out the procedures, we will focus on the high-level equipment that would be required to carry out such a protocol Materials As this chapter is focused on the development of an automated protocol for protoplast isolation and transformation, specialized equipment is required to carry out the procedure To provide the reader with an idea of the required setup, a schematic of the robotic platform developed in our lab is shown in Fig 1, which is based on microtiter plates The essential components of the system include a plate mover (Thermo Scientific™ Orbitor™ RS microplate mover) to transfer the plates between equipment, a plate shaker (Thermo Scientific™ Teleshake magnetic microplate shaker), a plate heater/ chiller (two InHeco peltier CPAC ultraflat HT 2-TEC microplate heater/chillers and one InHeco Multi-TEC controller), a largevolume liquid handler (Biotek MultiFlo FX multi-mode dispenser), a tip-based liquid handler (Agilent Bravo automated liquid handling system), and a plate reader (Biotek Synergy H1 hybrid multi-mode reader) In addition, the system must be housed in a Automated Protoplast Transformation 357 Fig Schematic of a robotic system for automated protoplast isolation, transformation, and screening The central component is the Orbitor RS microplate mover, which can transfer plates to any of the pieces of equipment, including one nest on the Agilent Bravo Protoplast isolation is primarily carried out using the Orbitor, MultiFlo FX reagent dispenser, the Teleshake plate shaker, and the InHeco peltier plate heater/chillers Transformation is carried out using all pieces of equipment, with the exception of the Synergy H1 plate reader, which is used for screening The Agilent Bravo is the primary piece of equipment used for transfer of protoplasts between the different containers, and also for dispensing reagents 18 h, plates are screened using the microplate reader to determine the efficiency of transformation contained environment that ensures sterility throughout the entire procedure Many system designs can be used to accomplish the methods described herein, thus the specific equipment will be annotated generally as described 2.1 Cell Culture BY-2 liquid culture media: Weigh 4.43 g Linsmaier and Skoog (LS) basal media (see Note 1), 30 g of sucrose, 200 mg, KH2PO4, and 200 μg of 2,4 dichlorophenoxyacetic acid (2,4-D) and add to a L beaker Add 900 mL of Milli-Q water and pH to 5.8 with 0.1 M KOH Make up to L with water and autoclave Store at  C for up to weeks BY-2 solid culture media: Add 1% agar to BY-2 liquid media, mix, and autoclave Pour plates prior to solidification of the agar BY-2 callus Wide bore 1.0 mL pipet tips 358 Scott C Lenaghan and C Neal Stewart Jr 2.2 Protoplast Isolation Digestion buffer: 0.4 M mannitol, mM CaCl2, 12 mM sodium acetate, pH 5.7 Protoplasting enzymes: Rohament CL (cellulase), Rohapect UF (pectinase), and Rohapect 10 L (pectinase/arabinase) (see Note 2) Complete protoplast isolation solution: Add 160 μL of Rohapect CL, 24 μL of Rohapect 10 L, and μL of Rohapect UF to 20 mL of digestion buffer and vortex Propidium iodide 2.3 PEG-Mediated Transformation Mmg solution: 0.4 M mannitol, 100 mM MgCl2, mM MES, pH 5.7 PEG solution: Dissolve g of PEG 4000 in 6.5 mL of Mmg solution and vortex W5 solution: 154 mM NaCl, 125 mM CaCl2, mM KCl, and mM MES, pH 5.7 Wide bore 1.0 mL pipet tips Deep 96-well, 1.2 mL plates Plasmid DNA: μg/μL, A260/280 > 1.8 3.1 Methods Cell Culture Propagate BY-2 callus on BY-2 solid culture media prior to establishment of cell suspension culture (see Note 3) Initiate liquid suspension culture by adding a single callus piece, >1 cm, to a 250 mL Erlenmeyer flask containing 100 mL of BY-2 liquid medium and seal with aluminum foil Incubate the culture at 28–30  C with constant shaking in the dark for days Subculture 2.0 mL of BY-2 cell suspension culture into 98 mL of BY-2 liquid medium in a 250 mL Erlenmeyer flask Incubate the culture at 28–30  C with constant shaking in the dark for 5–7 days (see Note 4) Collect cells for protoplast isolation by thoroughly mixing the flask prior to transfer of 6.0 mL of the culture into a 15 mL conical bottom tube Allow the culture to settle for ~10 Adjust the packed cell volume to 3.0 mL through removal of the supernatant (see Note 5) Vortex the 15 mL conical bottom tube to thoroughly mix the cell suspension, and transfer 500 μL to each well of a 6-well plate for protoplast isolation (see Note 6) Automated Protoplast Transformation 3.2 Protoplast Isolation 359 Load 6-well plate(s) containing the cell suspension cultures onto the plate mover (see Note 7) Add 2000 μL of complete protoplast isolation solution to each well of the 6-well plate using a large-volume liquid handler Move 6-well plate to plate heating station and incubate at 37  C for Move 6-well plate to plate shaker and shake at 500 rpm for Repeat steps and for a total of 18 loops (see Note 8) Move 6-well plate to plate chiller and incubate at  C for (see Note 9) Move 6-well plate to plate shaker and shake at 800 rpm for Move 6-well plate to tip-based liquid handler and transfer 70 μL of protoplasts from the 6-well plate to each well of a 96-well plate Pipet 70 μL of ethanol into the same wells previously loaded with protoplasts to fix the cells for counting Allow >10 at room temperature to fix and permeabilize the protoplasts Add 14 μL of propidium iodide (PI) to each of the fixed wells to label the nuclei of protoplasts The binding of PI to the nucleus enables the use of a plate reader to determine the number of protoplasts in each well when compared to a previously generated standard curve (see Note 10) 10 Move the 96-well plate to the plate reader and measure the fluorescence (536 nm excitation, 620 nm emission) of propidium iodide in the well All wells are compared to blank wells containing BY-2 liquid medium and propidium iodide 11 Compare the fluorescence reading with the previously generated standard curve to determine the concentration of protoplasts in each well (see Note 11) 3.3 PEG-Mediated Transformation Pipet 10 μL of plasmid DNA into each well of a deep 96-well plate (see Note 12) Move 6-well plate containing protoplasts to the plate shaker and shake at 800 rpm for Move 6-well plate to tip-based liquid handler and transfer 70 μL of protoplasts from the 6-well plate to each well of a deep 96-well plate (see Note 13) Transfer 70 μL of PEG solution into each well of the deep 96-well plate containing protoplasts (see Note 14) The final concentration of PEG in each well should be ~20%, depending on the volume of DNA added Move the deep 96-well plate to the plate shaker and shake at 1500 rpm for 30 s (see Note 15) 360 Scott C Lenaghan and C Neal Stewart Jr Incubate at room temperature without shaking for at least 20 to allow DNA to be taken up by the protoplasts Move deep 96-well plate to large-volume liquid handler and add 300 μL of W5 solution to each well and mix Add an additional 400 μL of W5 to each well (a 1:10 dilution of PEG in W5 is achieved in this protocol) (see Note 16) Move deep 96-well plate to plate shaker and shake at 1500 rpm for an additional to ensure complete mixing Incubate protoplasts for >1 h to allow protoplasts to settle to the bottom of the well Transfer 200 μL of transformed protoplasts to a 96-well fluorescent screening plate and incubate for 18–24 h (see Note 17) Move 96-well fluorescent screening plate to plate reader and measure the expression level of the fluorescent reporter gene (see Note 18) Notes Powdered media described in this work is typically purchased from Phytotech Laboratories® to prevent batch-to-batch variation of complex plant media In addition, while LS media is appropriate for culture of BY-2 tobacco cells, different media would be required for callus and cell suspension media from other plant species The use of low-cost food-grade enzymes is critical to highthroughput protoplast isolation [14, 15], as there is significant dead volume in the lines of liquid handlers, and typical lab-grade enzymes are costly Best results have been achieved using enzymes purchased from AB Enzymes For long-term maintenance of BY-2 cultures, it is recommended that callus be grown on solid media, as liquid cultures grow more rapidly It is also possible to cryopreserve BY-2 cultures and thaw as needed [16] Typically cell cultures are maintained in the dark; however, it is possible to obtain “green” cell suspension cultures in a variety of species by adjusting the media components and growing the cultures in the light While this is not possible for BY-2, it may be advantageous for other cell suspension cultures The packed cell volume used in this protocol has been validated to maximize digestion of BY-2 cells, and would need to be adjusted if using cell suspension cultures from another species or changing the enzymes used for digestion Wide-bore pipets or serological pipettes should be used to transfer the cells at this stage, as the cells are dense and will clog standard pipet tips Automated Protoplast Transformation 361 Numerous plate movers exist, and in robotic systems accomplish the automated movement of plates between the various pieces of equipment As this protocol is generally describing what would be required for automation, specific instrumentation will not be annotated It is possible to achieve both incubation and shaking on a single device; however, no such device is used in the current protocol In the event that shaking and incubation is accomplished on a single device, the cells would be incubated for ~3 h In order to prevent over-digestion of the cells, it is necessary to inactivate the enzymes by decreasing the temperature of the reaction If the enzymes are allowed to continuously digest the cells, the overall viability of the protoplasts will be significantly decreased 10 A variety of fluorescent dyes could be used to count the number of protoplasts, and it is possible to determine the protoplast viability using a combination of metabolically active dyes, such as fluorescein diacetate, in combination with propidium iodide 11 A standard curve should be generated manually using a fluorescent viability dye, in this case propidium iodide, to calibrate the plate reader Briefly, protoplasts should be isolated and concentrated to  106 protoplasts/mL in a volume of mL Protoplasts can then be fixed, stained with propidium iodide, and serially diluted across a plate By plotting the cell concentration vs fluorescence, a standard curve is generated that can be used to determine the number of protoplasts isolated in the automated procedure 12 In the current protocol, the plasmid DNA used is at a concentration of μg/mL, thus a total of 10 μg of DNA is used per transformation It is important to use at least this much DNA, but a smaller volume will not affect the transformation efficiency 13 The volume of protoplasts transferred, 70 μL, is the maximum volume of the tip-based liquid handler used in this protocol If another device is used, the volume could be adjusted accordingly 14 The PEG solution is highly viscous, thus it is important to slowly pipet the solution so that the volume is accurate This is especially important if using a liquid handler, where the pipetting speed should be adjusted to account for the increased viscosity 15 The PEG solution rapidly settles on the bottom of the well and thus complete mixing of protoplasts, DNA, and PEG requires additional mixing 362 Scott C Lenaghan and C Neal Stewart Jr 16 The PEG solution at the concentration required for transformation, 20%, is toxic to protoplasts after prolonged exposure, thus it is necessary to dilute the PEG after the transformation procedure is been completed 17 To increase the transformation efficiency, and to concentrate the protoplasts prior to screening, plates can be centrifuged at 100  g to pellet the protoplasts, followed by removal of the supernatant and resuspension in a smaller volume 18 In the current protocol, expression of a fluorescent protein reporter is used as the output; however, the output is not limited to fluorescent protein expression Acknowledgement This research was supported by Advanced Research Projects Agency-Energy (ARPA-E) Award No DE-AR0000313, with additional funding and assistance from ThermoFisher References Atkinson HJ, Lilley CJ, Urwin PE (2012) Strategies for transgenic nematode control in developed and developing world crops Curr Opin Biotechnol 23(2):251–256 Duke SO (2015) Perspectives on transgenic, herbicide-resistant crops in the United States almost 20 years after introduction Pest Manag Sci 71(5):652–657 Mir R, Zaman-Allah M, Sreenivasulu N, Trethowan R, Varshney R (2012) Integrated genomics, physiology and breeding approaches for improving drought tolerance in crops Theor Appl Genet 125(4):625–645 https:// doi.org/10.1007/s00122-012-1904-9 Hu H, Xiong L (2014) Genetic engineering and breeding of drought-resistant crops Annu Rev Plant Biol 65:715–741 Marco F, Bitria´n M, Carrasco P, Rajam MV, Alca´zar R, Tiburcio AF (2015) Genetic engineering strategies for abiotic stress tolerance in plants In: Plant Biology and Biotechnology Springer, New York, pp 579–609 Vanhercke T, El Tahchy A, Liu Q, Zhou XR, Shrestha P, Divi UK, Ral JP, Mansour MP, Nichols PD, James CN (2014) Metabolic engineering of biomass for high energy density: oilseed-like triacylglycerol yields from plant leaves Plant Biotechnol J 12(2):231–239 Baxter HL, Mazarei M, Labbe N, Kline LM, Cheng Q, Windham MT, Mann DG, Fu C, Ziebell A, Sykes RW (2014) Two-year field analysis of reduced recalcitrance transgenic switchgrass Plant Biotechnol J 12(7):914–924 Xing H-L, Dong L, Wang Z-P, Zhang H-Y, Han C-Y, Liu B, Wang X-C, Chen Q-J (2014) A CRISPR/Cas9 toolkit for multiplex genome editing in plants BMC Plant Biol 14 (1):327 Cao J, Yao D, Lin F, Jiang M (2014) PEG-mediated transient gene expression and silencing system in maize mesophyll protoplasts: a valuable tool for signal transduction study in maize Acta Physiol Plant 36 (5):1271–1281 10 Martinho C, Confraria A, Elias Carlos A, Crozet P, Rubio-Somoza I, Weigel D, BaenaGonza´lez E (2015) Dissection of miRNA pathways using Arabidopsis mesophyll protoplasts Mol Plant 8(2):261–275 https://doi.org/10 1016/j.molp.2014.10.003 11 Bart R, Chern M, Park C-J, Bartley L, Ronald PC (2006) A novel system for gene silencing using siRNAs in rice leaf and stem-derived protoplasts Plant Methods 2(1):13 12 Jiang F, Zhu J, Liu H-L (2013) Protoplasts: a useful research system for plant cell biology, especially dedifferentiation Protoplasma 250 (6):1231–1238 https://doi.org/10.1007/ s00709-013-0513-z 13 Mustafa NR, de Winter W, van Iren F, Verpoorte R (2011) Initiation, growth and Automated Protoplast Transformation cryopreservation of plant cell suspension cultures Nat Protoc 6(6):715–742 14 Dlugosz EM, Lenaghan SC, Stewart CN (2016) A robotic platform for highthroughput protoplast isolation and transformation JoVE J Vis Exp 115:e54300–e54300 15 Burris KP, Dlugosz EM, Collins AG, Stewart CN, Lenaghan SC (2016) Development of a 363 rapid, low-cost protoplast transfection system for switchgrass (Panicum virgatum L.) Plant Cell Rep 35(3):693–704 16 Kobayashi T, Niino T, Kobayashi M (2005) Simple cryopreservation protocol with an encapsulation technique for tobacco BY-2 suspension cell cultures Plant Biotechnol 22 (2):105–112 INDEX A CRISPR-Cpf1 (Cas12a) 11, 64, 245–255, 257–266, 338 CRISPR RNA (crRNA) 16, 48, 122, 204, 235, 245–255, 258, 270, 271, 273, 274, 279, 337, 338, 342, 345, 347, 348, 353 CsLOB1 235, 236, 238–240 Activation-induced cytidine deaminase (AIDs) 18, 19, 298–300, 302–306 Agrobacterium-mediated transformation 15, 149, 151, 152, 172, 177, 184, 189, 194–197, 236, 270, 272, 274–276, 313 Alternative end joining (altEj) 5, 7, 8, 10–13, 16, 17 Annealing at critical temperature-PCR (ACT-PCR) 27–32 Anthocyanins 91, 204, 212 Arabidopsis v, 7, 9–11, 16, 84, 87, 147–153, 158, 168, 172, 188, 199, 212, 235, 247, 249, 254, 255, 258, 259, 270, 271, 300, 312, 328 Artificial transcription factor 83 Automation .v, 355–362 Auxin binding protein (ABP1) 148, 149, 152, 153 dCas9-VP64 84, 85 Deaminases 16–18, 298–307 Decoding 33–42 Degenerate sequence decoding (DSD/ DSDecode) 34, 36–38, 41, 135 DNA repair v, 3–20, 48, 156, 286, 289 Double-strand breaks (DSBs) v, 4, 5, 7–17, 19, 27–30, 48, 75, 122, 147, 155–158, 184, 245, 246, 285, 286, 289, 292, 299, 312, 338 B E Base editors (BEs) 16, 17, 19, 20 Biolistic delivery 327–334 Brassica oleracea v, 155–169 Bright Yellow’ (BY-2) cell 356, 357, 360 Enzymatic digestion 71, 132, 198, 232, 252, 292, 303, 360, 361 C Callus 49–51, 56–58, 134, 140, 186, 194, 197, 204, 205, 208, 210, 212–214, 236, 264, 276, 286, 287, 303, 333, 334, 356, 357, 360 Carbon starved anther (CSA) 4, 5, 98, 99, 101, 102, 104, 105 Carrot 203–214 Cas9 nickase 7, 11, 246 Citrus v, 235–241 Composite plants 217, 218, 224, 225, 230 CRISPR-Cas9 .v, 11, 13, 17, 20, 27–32, 63–71, 75–81, 83, 97–106, 109, 121–140, 157, 175, 183–200, 203–214, 217–233, 235–241, 285–295, 299, 311–324, 327–334, 337–353, 355 D F Flavanone-3-hydroxylase (F3H) 204, 208, 211, 212, 214 Floral dip 149, 151–153 Francisella novicida Cpf1 (FnCpf1) 245, 259, 269–280, 341–345, 348, 353 G Gateway cloning 84, 122, 128, 131–132 Gene editing v, 10, 16, 20, 48, 147, 148, 171–181, 184, 203–214, 239, 338 Gene replacement v, 48, 121, 285–295 Genome editing 10, 19, 20, 27, 33, 35, 48, 63–65, 67, 75–81, 98, 110, 116, 121–140, 155, 156, 174, 178, 180, 183–200, 203, 204, 217–233, 245–247, 249, 251, 258, 259, 279, 285, 298, 306, 311–324, 327, 328, 337–353 Yiping Qi (ed.), Plant Genome Editing with CRISPR Systems: Methods and Protocols, Methods in Molecular Biology, vol 1917, https://doi.org/10.1007/978-1-4939-8991-1, © Springer Science+Business Media, LLC, part of Springer Nature 2019 365 PLANT GENOME EDITING 366 Index WITH CRISPR SYSTEMS Genome engineering 3–20, 83, 122, 147, 217, 218, 270, 312–315, 327 Golden Gate assembly 67, 68, 84, 91, 159, 164, 166 Gradient polyacrylamide gel electrophoresis (PAGE) 218, 220, 229, 232 Guide RNA (gRNA) 17, 48, 63, 83, 128, 148, 172, 188, 204, 218, 249, 259, 300, 312, 338 H Hairy root transformation 217, 219, 223–225 High-throughput screening 33, 197, 356 Homology-directed repair (HDR) .4, 7, 8, 10, 12–16, 48, 122, 204, 246, 285, 286, 289, 299, 312, 338 I Insertions and deletions (indels) 7, 8, 11, 12, 16–20, 27, 76, 122, 135–137, 156, 157, 159, 160, 172, 178, 204, 214, 239, 264, 265, 285–287, 289, 292, 294, 299, 322 Introns 13, 64, 66, 199, 265, 285–295 K Knockouts 76, 155–169, 174, 184, 187–189, 259, 328 L Lachnospiraceae bacterium ND2006 Cpf1 (LbCpf1) 11, 245, 251, 253, 254, 259, 261, 262, 264, 265, 270 Large chromosomal deletions 48–60 Large-scale 27 Lettuce 20, 328, 337–353 M Maize 18, 20, 87, 121–140, 204, 247, 253, 328 mCherry 148, 149, 152, 153 MCP-VP64 83 MicroRNAs (miRNAs) 109–111, 113, 116, 355 MS2 18, 83, 84 Multiplex genome editing .63–65, 67, 75–81, 259 Mutants 12, 13, 16, 19, 20, 27–33, 36, 37, 58, 102, 104, 105, 110, 122, 136–138, 140, 148, 149, 159, 160, 162, 167, 172, 194, 197, 198, 200, 204, 205, 210, 211, 232, 233, 259, 269, 280, 288, 314, 323, 328, 333 Mutant screening .27, 31, 334 N Next-generation sequencing (NGS) 239, 305, 306 Nicotiana benthamiana 312–315, 317–319, 324 Non-homologous end-joining (NHEJ) .4, 8, 11–13, 15, 16, 27, 48, 122, 147, 155–157, 179, 184, 204, 246, 251, 258, 285–289, 292, 294, 295, 299, 312, 338 O OsEPFL9 .259, 261–263 P PCR amplicon sequencing 172 Photoperiod-sensitive genic male sterility (PGMS) 97 Plant gene knockdown 247, 249, 255 Plant gene knockout 245–255 Plant transcriptional activation 83 Potato v, 20, 183, 184, 188, 194–197, 200, 269 Protoplasts 13, 18, 71, 218, 236, 266, 289, 291, 293, 328, 334, 338, 348–352, 355–357, 360–362 Protospacer adjacent motifs (PAMs) 18, 19, 27, 29, 30, 48, 58, 63, 67, 75, 87, 92, 111, 116, 122, 128, 147, 156, 157, 159, 163, 164, 168, 174, 184, 188, 194, 204, 208, 246, 251, 252, 258, 259, 270, 273, 286, 287, 291, 293, 305, 312, 314, 318, 322, 323, 338, 342 Protospacers 156, 157, 163, 164, 168, 184, 328, 330, 333 R Rapid 8, 27, 28, 34, 63, 128, 217, 220, 298, 360, 361 Ribonucleoprotein (RNP) 14, 20, 48, 76, 218, 327–334, 337–353 Ribozyme-gRNA-Ribozyme (RGR) 149 PLANT GENOME EDITING Rice 11–13, 18, 19, 29, 31, 33, 48–51, 55–58, 64–66, 71, 81, 97–99, 102–104, 106, 110, 114, 116, 122–124, 204, 235, 246, 247, 253, 254, 258–260, 262, 270, 286–289, 291–294, 300, 312, 328 RNA viruses 313 Robotics 356, 357, 361 S Sanger sequencing 33–41, 58, 89, 98, 102, 114, 117, 118, 164, 167, 168, 189, 193, 253, 255, 303, 306, 318, 319, 321–323 Single-guide RNA (sgRNA) 13, 16, 27, 48, 75, 87, 98, 110, 122, 123, 156, 158, 159, 184, 203, 235, 246, 279, 285, 298, 318, 327, 337 Single-strand breaks (SSBs) 4, 5, 7, 13–16, 63 Single strand conformation polymorphism (SSCP) 115–117 Single transcript unit (STU) 76, 78, 80–81 Site-directed nucleases (SDNs) 11, 13, 15, 20 Solanaceae 172, 298 Soybean 217–233, 246, 328 Streptococcus pyogenesCas9 (SpCas9) 48, 75, 121, 159, 184, 194, 245, 246, 341–345, 353 Superimposed chromatogram 34 T Target-AID 298–300, 302–306 Targeted modification .239, 312–315, 319 Targeted mutagenesis 11–13, 48, 58, 71, 75, 99, 110, 121, 123, 126, 128, WITH CRISPR SYSTEMS Index 367 134–136, 149, 157, 166, 184, 194, 197–198, 218, 259, 269–280, 322, 328 Targeted nucleotide substitutions 298, 300 T7 endonuclease (T7E1) assay 33, 57, 58, 172, 178–181, 194, 198, 315, 321, 323 Tissue culture 50–51, 57, 105, 184, 203, 213, 217, 231, 237, 266, 294, 313–315, 330, 333, 334 Tobacco 11, 15, 246, 259, 269–280, 328, 356, 360 Tobacco rattle virus (TRV) 313–315, 317–319, 321–324 Tomato v, 15, 171–181, 298–307, 312 Transformation 15, 56, 65, 102, 114, 122, 153, 157, 177, 187, 203, 217, 235, 253, 262, 272, 289, 303, 313, 327, 350, 356 Transgene-free 102, 104, 136, 140, 148, 159, 160, 162, 167, 259, 264–266, 313, 328 tRNA 63–71, 246 Two-line hybrid rice 97, 98 V Vector .13, 41, 51, 65, 76, 84, 100, 110, 122, 148, 157, 172, 184, 204, 218, 236, 247, 259, 269, 286, 300, 313, 327, 341 W Wheat v, 18, 20, 269, 293, 312, 327–334 ... MD, USA ISSN 106 4-3 745 ISSN 194 0-6 029 (electronic) Methods in Molecular Biology ISBN 97 8-1 -4 93 9-8 99 0-4 ISBN 97 8-1 -4 93 9-8 99 1-1 (eBook) https://doi.org/10.1007/97 8-1 -4 93 9-8 99 1-1 Library of Congress... analysis of CRISPR- induced mutations (Chapters and 3), multiplexed CRISPR- Cas9 systems (Chapters 4–7), CRISPR- Cas9 editing in monocots (e.g., rice and maize; Chapters 8–10), CRISPR- Cas9 editing in... diverse kinds of Yiping Qi (ed.), Plant Genome Editing with CRISPR Systems: Methods and Protocols, Methods in Molecular Biology, vol 1917, https://doi.org/10.1007/97 8-1 -4 93 9-8 99 1-1 _1, © Springer

Ngày đăng: 08/05/2020, 06:39

Từ khóa liên quan

Mục lục

  • Preface

  • Contents

  • Contributors

  • Part I: Review on Plant DNA Repair and Genome Editing

    • Chapter 1: Plant DNA Repair Pathways and Their Applications in Genome Engineering

      • 1 Plant DNA Repair and Recombination Machineries

      • 2 Role of DNA Recombination and Repair Machineries in Plant Development

      • 3 Targeted Mutagenesis and Insertion Mediated by NHEJ Repair Pathways

      • 4 HDR-Mediated Sequence Replacement with Synthetic Oligodeoxyribonucleotide (ODN) Donors

      • 5 Targeted Insertion of Large Size Donor Sequence Through Homologous Recombination

      • 6 Site-Directed Base Editing

      • 7 Perspectives

      • References

      • Part II: CRISPR Design and Mutation Analysis

        • Chapter 2: Rapid Screening of CRISPR/Cas9-Induced Mutants Using the ACT-PCR Method

          • 1 Introduction

          • 2 Materials

          • 3 Methods

            • 3.1 Primer Design

            • 3.2 Detection of the Critical Annealing Temperature by Preliminary Gradient PCR

            • 3.3 Mutant Screening

            • 4 Notes

            • References

            • Chapter 3: Decoding Sanger Sequencing Chromatograms from CRISPR-Induced Mutations

              • 1 Introduction

              • 2 Materials

Tài liệu cùng người dùng

Tài liệu liên quan